Lymphedema: Complete Medical and Surgical Management [1 ed.] 9781626236714

Lymphedema: Complete Surgical and Medical Management is one of the first books to cover the state of the field, encompas

1,324 116 238MB

English Pages 654 [657] Year 2015

Report DMCA / Copyright

DOWNLOAD FILE

Polecaj historie

Lymphedema: Complete Medical and Surgical Management [1 ed.]
 9781626236714

Table of contents :
Dedications
Contributors
Preface
Acknowledgments
Contents
Part I: Current Concepts in Lymphedema
1 Lymphedema: Lack of Solutions to a Clinical Problem • Manish C. Champaneria, Peter C. Neligan
2 Lymphedema and Its Impact on Quality of Life • Jane M. Armer, Jennifer M. Hulett, Janice N. Cormier, Bob R. Stewart, Ausanee Wanchai, Kate D. Cromwell
3 Quality of Life Measurement Instruments • Vaughan Keeley
Part II: Anatomy, Physiology, and Lymphangiogenesis
4 Embryology • Sandro Michelini, Miguel Amore, Lucia Tapia, Gisela Romina Pattarone, Diego Mercado, Jeremy S. Torrisi, Ira L. Savetsky, Jason C. Gardenier, Babak J. Mehrara
5 Changing Concepts in Lymphatic Pathways • Wei-Ren Pan
6 Applied Anatomy • Miguel Amore, Lucia Tapia, Gisela Romina Pattarone, Diego Mercado
7 Lymphangiogenesis • Jeremy S. Torrisi, Ira L. Savetsky, Jason C. Gardenier, Babak J. Mehrara
8 Impact of Genetics on Lymphangiogenesis • Kristiana Gordon, Pia Ostergaard
9 Relationship Between Fat Tissue and Lymphangiogenesis • Mauro Andrade
10 Lymphatic Malformations • Sandro Michelini, Alessandro Fiorentino, Marco Cardone
11 Immune Regulation by the Peripheral Lymphatics • David G. Hancock
Part III: Pathophysiology and Clinical Presentation
12 Pathophysiology of Primary Lymphedema • Byung-Boong Lee, James Laredo
13 Pathophysiology of Secondary Lymphedema • Etelka Földi
14 Dermatologic Implications of Secondary Lymphedema of the Lower Leg • Terence Ryan, S.R. Narahari, B. Vijaya, Madhur Guruprasad Aggithaya
15 Filaria • Gurusamy Manokaran, Rajiv Agarwal, Devisha Agarwal
16 Lymphedema in Pediatric Patients • Cristobal Miguel Papendieck
17 Combined Lymphatic and Venous Failure: Phlebolymphedema • Audra A. Duncan
18 Lymphedema Risk Factors in Breast Cancer • Swetha Kambhampati, Stanley Rockson
19 Sentinel Lymph Node Biopsy Outcomes • Janice N. Cormier, Kate D. Cromwell, Jane M. Armer, Merrick I. Ross
Part IV: Diagnosis of Lymphedema
20 Causes and Classification of Lymphatic Disorders • Swetha Kambhampati, Stanley Rockson
21 Basic Approaches to the Diagnosis of Lymphedema: Clinicians’ Perspective • Yener Demirtas, Baris Yigit
22 Biomarkers • Kerstin Buttler, Jörg Wilting
23 Clinical Staging of Lymphedema • Sandro Michelini, Marco Cardone, Alessandro Fiorentino
24 Measuring Methods • Peter C. Neligan
25 Hydromechanics of Intercellular Fluid and Lymph • Waldemar Olszewski, Marzanna T. Zaleska
26 Radionuclide Lymphoscintigraphy • Byung-Boong Lee, James Laredo
27 Indocyanine Green Lymphography • Mitsunaga Narushima, Takumi Yamamoto, Isao Koshima
28 Magnetic Resonance Lymphangiography • Lee M. Mitsumori
Part V: Current Treatment of Lymphedema
29 Conservative Treatments for Lymphedema • Neil B. Piller
30 Pharmacologic Treatment of Lymphedema • Kathleen Wang
31 Excisional Approaches for the Treatment of Lymphedema • Manish C. Champaneria, Peter C. Neligan
32 Liposuction • Håkan Brorson
33 Multiple Lymphaticovenous Anastomoses and Multiple Lymphatic-Venous-Lymphatic Anastomoses • Corrado Cesare Campisi, Melissa Ryan, Corradino Campisi
34 Lymphatic Grafts • Rüdiger G.H. Baumeister
35 Lymphaticovenular Anastomosis • Isao Koshima, Mitsunobu Harima
36 Vascularized Lymph Node Transfer • Corinne Becker
37 Reverse Lymphatic Mapping • Joseph H. Dayan, Mark L. Smith
38 Local Flaps and Vascularized Lymph Node Transfer • Joshua Levine, Corinne Becker
39 Transverse Myocutaneous Gracilis With Vascularized Lymph Node Transfer • Sinikka Suominen, Maija Kolehmainen
40 Lymphatic Microsurgical Preventing Healing Approach Concept • Francesco Boccardo
41 Combined Surgical Treatment for Breast Cancer–Related Lymphedema • Jaume Masia
42 Interventional Timing • Peter C. Neligan, Jaume Masia
43 Lymphedema Complications and Their Treatment • Arin K. Greene, Reid A. Maclellan
44 Treatment of Lymphedema in India • Rajiv Agarwal, Devisha Agarwal
45 Surgical Treatment in China • Ningfei Liu
Part VI: Research and Future Directions
46 Animal Models of Lymphedema • Swapna Ghanta, Daniel A. Cuzzone, Babak J. Mehrara
47 Harnessing Stem Cell–Mediated Lymphangiogenesis for the Treatment of Lymphedema • Ramin Shayan, Geraldine Mitchell, Anthony Penington
48 Establishment of a Lymphedema Framework • Christine Moffatt, Susie Murray
Credits
Index

Citation preview

Lymphedema Complete Medical and Surgical Management

K23349_00_Neligan_FM_m6_cl_i-xxii.indd 1

5/29/2015 9:24:23 AM

K23349_00_Neligan_FM_m6_cl_i-xxii.indd 2

5/29/2015 9:24:24 AM

Lymphedema Complete Medical and Surgical Management

Editors Peter C. Neligan, MB, FRCS(I), FRCSC, FACS Professor of Surgery, Division of Plastic Surgery, University of Washington, Seattle, Washington

Jaume Masia, MD, PhD

Chief, Plastic Surgery Department, Hospital de la Santa Creu i Sant Pau; Professor of Plastic Surgery, School of Medicine at Universitat Autònoma de Barcelona; Director, Microsurgery and Breast Reconstructive Unit, Clinica Planas, Barcelona, Spain

Neil B. Piller, BS, PhD, FACP

Professor and Director, Lymphoedema Research Unit, Department of Surgery, Flinders University School of Medicine, Bedford Park, Adelaide, South Australia, Australia

Illustrated by Cassio Lynm

K23349_00_Neligan_FM_m8_cl_i-xxii.indd 3

5/29/2015 1:53:44 PM

Director, Editorial Services: Mary Jo Casey International Production Director: Andreas Schabert International Marketing Director: Fiona Henderson International Sales Director: Louisa Turrell Director of Sales, North America: Mike Roseman Senior Vice President and Chief Operating Officer: Sarah Vanderbilt President: Brian D. Scanlan Library of Congress Cataloging-in-Publication Data is available from the publisher upon request.

Important note: Medicine is an ever-changing science undergoing continual development. Research and clinical experience are continually expanding our knowledge, in particular our knowledge of proper treatment and drug therapy. Insofar as this book mentions any dosage or application, readers may rest assured that the authors, editors, and publishers have made every effort to ensure that such references are in accordance with the state of knowledge at the time of production of the book. Nevertheless, this does not involve, imply, or express any guarantee or responsibility on the part of the publishers in respect to any dosage instructions and forms of applications stated in the book. Every user is requested to examine carefully the manufacturers’ leaflets accompanying each drug and to check, if necessary in consultation with a physician or specialist, whether the dosage schedules mentioned therein or the contraindications stated by the manufacturers differ from the statements made in the present book. Such examination is particularly important with drugs that are either rarely used or have been newly released on the market. Every dosage schedule or every form of application used is entirely at the user’s own risk and responsibility. The authors and publishers request every user to report to the publishers any discrepancies or inaccuracies noticed. If errors in this work are found after publication, errata will be posted at www.thieme.com on the product description page. Some of the product names, patents, and registered designs referred to in this book are in fact registered trademarks or proprietary names even though specific reference to this fact is not always made in the text. Therefore, the appearance of a name without designation as proprietary is not to be construed as a representation by the publisher that it is in the public domain.

© 2016 Thieme Medical Publishers, Inc. Thieme Publishers New York 333 Seventh Avenue, New York, NY 10001 USA +1 800 782 3488, [email protected] Thieme Publishers Stuttgart Rüdigerstrasse 14, 70469 Stuttgart, Germany +49 [0]711 8931 421, [email protected] Thieme Publishers Delhi A-12, Second Floor, Sector-2, Noida-201301 Uttar Pradesh, India +91 120 45 566 00, [email protected] Thieme Publishers Rio de Janeiro, Thieme Publicações Ltda. Edifício Rodolpho de Paoli, 25º andar Av. Nilo Peçanha, 50 – Sala 2508 Rio de Janeiro 20020-906, Brasil +55 21 3172 2297 eISBN 978-1-62623-753-7

This book, including all parts thereof, is legally protected by copyright. Any use, exploitation, or commercialization outside the narrow limits set by copyright legislation without the publisher’s consent is illegal and liable to prosecution. This applies in particular to photostat reproduction, copying, mimeographing or duplication of any kind, translating, preparation of microfilms, and electronic data processing and storage.

This book is dedicated to my wife, Gabrielle, who, as always, is my staunch supporter



P.C.N.

This book is dedicated to my family for their unconditional love and support, which allowed me to achieve my professional dreams



J.M.

To my fantastic wife, loving family, and caring friends for being there when I needed them, and for their enduring enthusiasm and support



N.B.P.

K23349_00_Neligan_FM_m6_cl_i-xxii.indd 5

5/29/2015 9:24:24 AM

EXECUTIVE EDITOR  Sue Hodgson SENIOR PROJECT EDITING MANAGER  Carolyn Reich SENIOR DEVELOPMENTAL EDITOR  Megan Fennell GRAPHICS MANAGER  Brett Stone DIRECTOR OF ILLUSTRATION AND DESIGN  Brenda Bunch MANAGING EDITOR  Suzanne Wakefield PROJECT MANAGER  Idelle Winer PRODUCTION  Chris Lane, Debra Clark, Susan Trail, Madonna Gauding PROOFREADER  Linda Maulin INDEXER  Matthew White

K23349_00_Neligan_FM_m6_cl_i-xxii.indd 6

5/29/2015 9:24:25 AM

C ontributors Devisha Agarwal, MBBS (Std) Medical Student, GSVM Medical College, Kanpur, Uttar Pradesh, India

Corinne Becker, MD Lecturer, Department of Thoracic Surgery, Hôpital Européen–Georges Pompidou; Surgeon, American Hospital of Paris, Paris, France

Rajiv Agarwal, MCh, FRCS Professor and Head, Department of Plastic Surgery and Burns, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow-Uttar Pradesh, India

Francesco Boccardo, MD, PhD Associate Professor of Surgery, Unit of Lymphatic Surgery, Department of Surgery; Director, Clinic for Medical Oncology, IRCCS University Hospital San Martino, National Institute for Cancer Research (IST), Genoa, Italy

Madhur Guruprasad Aggithaya, MD Institute of Applied Dermatology, Kasaragod, Kerala, India

Håkan Brorson, MD, PhD Associate Professor, Department of Clinical Sciences, Lund University; Department of Plastic and Reconstructive Surgery, Skåne University Hospital, Malmö, Sweden; Professor, Esculera de Graduados, Asociación Médica Argentina, Buenos Aires, Argentina

Miguel Amore, MD, PhD, FACS Director of Vascular Anatomy Laboratory; III Chair of Anatomy, Department of Anatomy, Buenos Aires University; Surgical Staff, Unit of Phlebology and Lymphology, Central Military Hospital, Buenos Aires, Argentina

Kerstin Buttler, MD Professor, Institute of Anatomy and Cell Biology, University Medical Center Göttingen, Göttingen, Germany

Mauro Andrade, MD, PhD Associate Professor of Surgery, Department of Surgery, University of São Paulo Medical School, São Paulo, Brazil

Corradino Campisi, MD, PhD, FACS University Department of Surgery (DISC), Section of Lymphology and Microsurgery, Operative Unit of Lymphatic Surgery, IRCCS University Hospital San Martino, National Institute for Cancer Research (IST), Genoa, Italy

Jane M. Armer, PhD, RN, CLT, FAAN Professor, Sinclair School of Nursing, University of Missouri; Director, Department of Nursing Research, Ellis Fischel Cancer Center; Director, American Lymphedema Framework Project, Columbia, Missouri

Corrado Cesare Campisi, MD, PhD, RAS-ACS Surgeon, Division of Plastic, Reconstructive and Aesthetic Surgery, Department of Surgery (DISC), IRCCS University Hospital San Martino (IST), Genoa; Surgeon, Division of Experimental and Microsurgery, Department of Surgery, IRCCS San Matteo University Hospital Foundation, Pavia, Italy

Rüdiger G.H. Baumeister, MD, PhD Professor, Former Head, Department of Surgery, Campus Grosshadern, Division of Plastic, Hand, and Micro-Surgery, Ludwig Maximilians University; Consultant in Lymphology, Department of Surgery, Chirurgische Klinik München-Bogenhausen, Munich, Germany vii

K23349_00_Neligan_FM_m6_cl_i-xxii.indd 7

5/29/2015 9:24:25 AM

viii

Contributors

Marco Cardone, MD Department of Rehabilitation Medicine, San Giovanni Battista Hospital, Rome, Italy

Etelka Földi, MD Professor, Center for Lymphology, Földiklinik, Hinterzarten-Baden-Württemberg, Germany

Manish C. Champaneria, MD Plastic and Reconstructive Surgeon, Department of Surgery, PeaceHealth Southwest Medical Center, Vancouver, Washington

Jason C. Gardenier, MD Research Fellow, Department of Surgery, Division of Plastic and Reconstructive Surgery, Memorial Sloan Kettering Cancer Center, New York, New York

Janice N. Cormier, MD, MPH, FACS Professor of Surgery, Department of Surgical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas Kate D. Cromwell, MS, MPH Clinical Studies Coordinator, Department of Surgical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas Daniel A. Cuzzone, MD Resident Research Fellow, Department of Surgery, Division of Plastic and Reconstructive Surgery, Memorial Sloan Kettering Cancer Center; Resident, Department of Plastic Surgery, Institute of Reconstructive Plastic Surgery, New York University Langone Medical Center, New York, New York Joseph H. Dayan, MD Assistant Professor, Department of Surgery, Division of Plastic and Reconstructive Surgery, Memorial Sloan Kettering Cancer Center, New York, New York Yener Demirtas, MD Associate Professor, Lymphest Plastic Surgery Clinic, Istanbul, Turkey Audra A. Duncan, MD Professor of Surgery, Mayo Clinic, Rochester, Minnesota Alessandro Fiorentino, MD Department of Vascular Surgery, Università Cattolica del Sacro Cuore, Policlinico Agostino Gemelli, Rome, Italy

K23349_00_Neligan_FM_m6_cl_i-xxii.indd 8

Swapna Ghanta, MD Research Fellow, Department of Surgery, Division of Plastic and Reconstructive Surgery, Memorial Sloan Kettering Cancer Center, New York, New York Kristiana Gordon, MBBS, MRCP, CLT, MD (Res) Honorary Senior Lecturer, Cardiovascular and Cell Sciences Research Institute, St. George’s University of London; Consultant, Department of Dermatology and Lymphovascular Medicine, St. George’s Hospital, London, United Kingdom Arin K. Greene, MD, MMSc Co-director, Lymphedema Program, Department of Plastic and Oral Surgery, Boston Children’s Hospital; Associate Professor of Surgery, Harvard Medical School, Boston, Massachusetts David G. Hancock, PhD Lymphoedema Research Unit, Department of Surgery, Flinders University School of Medicine, Bedford Park, Adelaide, South Australia, Australia Mitsunobu Harima, MD Department of Clinical Pharmacology, Niigata University of Pharmacy and Applied Life Sciences, Niigata, Japan Jennifer M. Hulett, PhD(c), APRN, FNP-BC Doctoral Candidate, Sinclair School of Nursing, University of Missouri, Columbia, Missouri Swetha Kambhampati, MD Resident in Internal Medicine, The Johns Hopkins Hospital, Baltimore, Maryland

5/29/2015 9:24:25 AM

Contributors

Vaughan Keeley, PhD, FRCP Honorary Associate Professor, Department of Palliative Medicine, University of Nottingham School of Medicine, Nottingham; Consultant in Palliative Medicine/Lymphoedema, Derby Teaching Hospitals, NHS Foundation Trust, Derby, United Kingdom

Jaume Masia, MD, PhD Chief, Plastic Surgery Department, Hospital de la Santa Creu i Sant Pau; Professor of Plastic Surgery, School of Medicine at Universitat Autònoma de Barcelona; Director, Microsurgery and Breast Reconstructive Unit, Clinica Planas, Barcelona, Spain

Maija Kolehmainen, MD Department of Plastic Surgery, Helsinki University Central Hospital, Helsinki, Finland

Babak J. Mehrara, MD, FACS Director, Lymphatic Research Laboratory, Department of Surgery, Division of Plastic and Reconstructive Surgery, Memorial Sloan Kettering Cancer Center; Professor of Plastic Surgery, Weill Cornell University Medical Center, New York, New York

Isao Koshima, MD Professor and Chief, Department of Plastic, Reconstructive and Aesthetic Surgery, The University of Tokyo Hospital, Tokyo, Japan James Laredo, MD, PhD, FACS Associate Professor of Surgery, Department of Surgery, Division of Vascular Surgery, George Washington University Medical Center, Washington, DC Byung-Boong Lee, MD, PhD, FACS Professor of Surgery, Department of Surgery, George Washington University, Washington, DC Joshua Levine, MD Director of Surgical Services, Department of Plastic and Reconstructive Surgery, New York Eye and Ear Infirmary of Mount Sinai, New York, New York Ningfei Liu, MD, PhD Professor of Plastic Surgery, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China Reid A. Maclellan, MD, MMSc Instructor of Surgery, Department of Plastic and Oral Surgery, Lymphedema Program, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts Gurusamy Manokaran, MD Senior Consultant Plastic Surgeon and Lymphologist, Apollo Hospital, Chennai, India

K23349_00_Neligan_FM_m6_cl_i-xxii.indd 9

ix

Diego Mercado, MD Institute for Immunological Research, University of Cartagena, Cartagena, Colombia Sandro Michelini, MD Chief of Vascular Rehabilitation, San Giovanni Battista Hospital; Past President, European Society of Lymphology; President, Italian Lymphoedema Framework; President-Elect, Italian Society of Phlebolymphology, Rome, Italy Geraldine Mitchell, PhD Faculty of Health Sciences, Australian Catholic University, Melbourne, Victoria, Australia Lee M. Mitsumori, MD, MSBE Affiliate Associate Professor, Department of Radiology, University of Washington, Seattle, Washington; Physician, Department of Radiology, Straub Clinic and Hospital, Honolulu, Hawaii Christine Moffatt, CBE, FRCN, PhD, MA, RGN, DN Professor of Clinical Nursing Research, School of Health Sciences, The University of Nottingham, Nottingham, United Kingdom Susie Murray MA, RGN, RHV (retired) Project Manager, International Lymphoedema Framework, London, United Kingdom S.R. Narahari, MD, DVD Chair and Director, Institute of Applied Dermatology, Kasaragod, Kerala, India

5/29/2015 9:24:25 AM

x

Contributors

Mitsunaga Narushima, MD Assistant Professor, Department of Plastic and Reconstructive Surgery, University of Tokyo, Tokyo, Japan Peter C. Neligan, MB, FRCS(I), FRCSC, FACS Professor of Surgery, Division of Plastic Surgery, University of Washington, Seattle, Washington Waldemar Olszewski, MD, PhD Professor of Surgery, Medical Research Center, Polish Academy of Sciences, Central Clinical Hospital, Ministry of Internal Affairs, Warsaw, Poland Pia Ostergaard, PhD Senior Research Fellow, Lymphovascular Research Unit, Division of Cardiovascular and Cell Sciences, St. George’s University of London, London, United Kingdom Wei-Ren Pan, MD, PhD Professor, Department of Anatomy, Xuzhou Medical College, Xuzhou, Jiangsu, China Cristobal Miguel Papendieck, MD, FACS Department of Pediatric Surgery, Deutsches Hospital, Buenos Aires, Argentina Gisela Romina Pattarone, MD Department of Medicine, University of Buenos Aires School of Medicine, Buenos Aires, Argentina Anthony Penington, MB, BS, FRACS Jigsaw Professor of Pediatric and Maxillofacial Surgery, The Royal Children’s Hospital, Melbourne, Victoria, Australia Neil B. Piller, BS, PhD, FACP Professor and Director, Lymphoedema Research Unit, Department of Surgery, Flinders University School of Medicine, Bedford Park, Adelaide, South Australia, Australia

Stanley Rockson, MD, FACP, FACC Allan and Tina Neill Professor of Lymphatic Research and Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine; Director, Stanford Center for Lymphatic and Venous Disorders, Stanford University Hospital and Clinics, Stanford, California Merrick I. Ross, MD Chief, Melanoma Section, Charles C. McBride Distinguished Professorship in Surgical Oncology, Department of Surgical Oncology, Division of Surgery, University of Texas M.D. Anderson Cancer Center, Houston, Texas Melissa Ryan, PhD, PgDips Clinical Psychologist, Department of Surgery, Section of Lymphology and Microsurgery, Operative Unit of Lymphatic Surgery, IRCCS University Hospital San Martino, Genoa, Italy Terence Ryan, DM, FRCP, KStJ Emeritus Professor, Department of Dermatology, Green Templeton College, Oxford University, Oxford, United Kingdom Ira L. Savetsky, MD Research Fellow, Department of Surgery, Division of Plastic and Reconstructive Surgery, Memorial Sloan Kettering Cancer Center; Resident, Department of Surgery, New York University Langone Medical Center, New York, New York Ramin Shayan, MB, BS, PhD, FRACS Jack Brockhoff Reconstructive Plastic Surgery Research Unit, Department of Anatomy and Cell Biology, The University of Melbourne, Parkville, Victoria, Australia Mark L. Smith, MD Division of Plastic and Reconstructive Surgery, Department of Surgery, Beth Israel Medical Center, New York, New York Bob R. Stewart, EdD Professor Emeritus, College of Education; Adjunct Professor, Sinclair School of Nursing, University of Missouri, Columbia, Missouri

K23349_00_Neligan_FM_m6_cl_i-xxii.indd 10

5/29/2015 9:24:25 AM

Contributors

Sinikka Suominen, MD, PhD Adjunct Professor and Department Chief, Clinic of Plastic Surgery, Helsinki University Hospital, Helsinki, Finland

Kathleen Wang, B. Psych (Hon), MD candidate Research Officer, Lymphoedema Research Unit, Flinders University School of Medicine, Bedford Park, Adelaide, South Australia, Australia

Lucia Tapia, MD Department of Medicine, University of Buenos Aires School of Medicine, Buenos Aires, Argentina

Jörg Wilting, MD Professor, Institute of Anatomy and Cell Biology, Göttingen University Medical Center, Göttingen, Germany

Jeremy S. Torrisi, BA Research Assistant, Department of Surgery, Division of Plastic and Reconstructive Surgery, Memorial Sloan Kettering Cancer Center, New York, New York

Takumi Yamamoto, MD Department of Plastic and Reconstructive Surgery, University of Tokyo, Tokyo, Japan

B. Vijaya, MD Professor of Pathology, JSS Medical College of JSS University, Mysore, Karnataka, India Ausanee Wanchai, PhD, RN Nursing Instructor, Deputy Director for Academic Service and Research, Boromarajonani College of Nursing, Buddhachinaraj Hospital, Phitsanulok, Thailand

K23349_00_Neligan_FM_m6_cl_i-xxii.indd 11

xi

Baris Yigit, MD Associate Professor, Düzce University, Department of Plastic Surgery, Düzce, Turkey Marzanna T. Zaleska, MD Medical Research Center, Polish Academy of Sciences, Department of Vascular and General Surgery and Radiology, Central Clinical Hospital, Ministry of Internal Affairs, Warsaw, Poland

5/29/2015 9:24:25 AM

K23349_00_Neligan_FM_m6_cl_i-xxii.indd 12

5/29/2015 9:24:25 AM

P reface Lymphedema is a significant global problem, and its incidence will increase with a population that is living longer. In addition, because of the interrelationship between the lymphatic system and adipose tissue, the obesity epidemic has led to a rapid rise in this complication. Lymphedema is unique in that it is the end result of myriad conditions affecting the blood, tissue, and lymphatic system, ranging from congenital anomalies to parasitic infestations to postcancer treatment sequelae and soft tissue trauma. Although we know a great deal about lymphedema in terms of its causes and consequences, we still know far too little to be able to propose a comprehensive targeted and sequenced treatment plan. So much work remains to be undertaken, and our knowledge base must be deepened and widened in this area. Lymphedema has been one of those conditions that most people do not want to acknowledge or treat. This is probably because we do not yet have a strong, standardized assessment regimen, an ability (and often willingness) to undertake an accurate differential diagnosis (separating lymphedema from other reasons for swelling), and a good objective knowledge of how best to sequence and target treatment. We still lack strong evidence as to which treatments are best, as well as an uncertainty about when to move from conservative to surgical strategies when conservative approaches fail, or even whether it is better to start with a surgical intervention (such as anastomoses) in the first place. Fortunately, there is a core group of practitioners who maintain their interest in this broad-based disorder, and in recent years, surgeons have joined them in recognition that surgical options after the failure of conservative treatment (or at times surgery may be added to conservative treatment early in the development of lymphedema) can offer improved management of the condition and thus improved quality of life for the at-risk group as well as those who already have lymphedema. In this book we highlight the surgical group, who have added a new dimension to our approach. We have tried to be rational, and where possible, evidence-based in our approach to lymphedema, although we acknowledge that at times the evidence is not strong or is based only on clinical judgments (the experience of experts). The fact that the symptoms and consequences of lymphedema often have a lifelong impact on the quality of survival of our patients must be dealt with in a balanced and holistic manner. For that reason, the book is divided into several parts. We have recruited the brightest minds and those with the greatest experience and expertise in the field, and put together a comprehensive, balanced, and considered approach to the condition. Lymphologists, therapists, and surgeons were asked to give their accurate (evidence-based and knowledge-based) appraisal of the problem. The result is a comprehensive catalog of where we are, what we know, and where we should go. xiii

K23349_00_Neligan_FM_m6_cl_i-xxii.indd 13

5/29/2015 9:24:25 AM

xiv

Preface

Part I outlines current concepts as a place to start. In Part II we discuss the anatomy and physiology of lymphedema and explore lymphangiogenesis. Embryology is also important to our understanding of the lymphatic system, particularly to our comprehension of lymphatic malformations. Furthermore, as our genetic knowledge deepens, additional genetic abnormalities are recognized. Here we examine their impact on these patients and their families. Finally, in this section the role of the lymphatics is addressed, as well as the general and specific aspects of immune system function, and the consequences when it fails. Part III examines in detail the pathophysiology of both primary and secondary lymphedema. Filariasis, the most common worldwide cause of lymphedema with over 1.4 billion at risk, is covered in a separate chapter. We also examine the impact of venous failure on lymphedema and how this and an often combined lymphatic failure affects adults as well as children. We tackle risk factors for lymphedema in patients with breast and other cancers and examine the role of sentinel node sampling in their management. Part IV emphasizes the fact that accurate diagnosis of lymphedema is vitally important; this is dealt with in its own section. Internationally accepted classification recommendations for lymphedema are discussed, because these systems offer a very useful tool to categorize clinical findings. The various assessment techniques, measurement options, imaging methods, and staging systems are reviewed, as well as lymph pressure and flow measurement using traditional and new techniques. We have tried to include all aspects of treatment. In Part V seventeen chapters have been assembled to cover every facet. These chapters not only include conservative approaches but also all of the surgical approaches currently being used around the world, together with recommendations for when and where they might be appropriate. One of the most important points emphasized in this section is how to recognize and treat the complications and comorbidities that are inevitable with this disease. In Part VI the book concludes with a review and critical appraisal of current research. This inevitably leads to a discussion of future directions. As mentioned at the outset, so much is unknown about lymphedema that there is a huge amount of work to be done. We explore several likely approaches that may be adopted. Lymphedema is presently of intense interest to investigators, and much research is being done in many areas, including genetics, conservative treatment and management, and surgical approaches, as well as the best strategies for early detection of lymphatic insufficiencies that can lead to lymphedema. Predictably, much of the information in this book will change in the coming years as we learn more and further understand the disease. However, it is our hope that this book will form the basis for new knowledge, enhance surgeons’ ability to explore innovative approaches, and lead all to provide better outcomes for patients at risk for and with lymphedema. Peter C. Neligan Seattle, Washington Jaume Masia Barcelona, Spain Neil B. Piller Bedford Park, Adelaide, South Australia, Australia

K23349_00_Neligan_FM_m6_cl_i-xxii.indd 14

5/29/2015 9:24:25 AM

A cknowledgments I would like first of all to acknowledge my two co-editors, Jaume and Neil, as well as the contributing authors who gave freely of their time and expertise to make this book possible. I am also indebted to my residents and fellows for always keeping me on my toes. My partners at the University of Washington are always supportive and encouraging, and I am grateful to them for that. It has been a pleasure working with the team at CRC Press led by Sue Hodgson, who, as always, keeps the project going and is constantly available to help and advise. Megan Fennell has done a stellar job, as has Idelle Winer. Brenda Brunch has directed the artwork, and Cassio Lynm has worked tirelessly with the contributing authors to make the illustrations not only look great but accurate. This book is a credit to all of them. P.C.N.

There are many people to whom I am indebted for making this publication possible. My heartfelt thanks to Peter and Neil for their expertise, enthusiasm, and honesty, which have been key factors in making this book a valuable resource for people wanting to learn more about lymphedema and to advance its treatment. I am sincerely grateful to the many colleagues and friends for their close collaboration and excellent contributions. Also to be thanked are the teams at Sant Pau University Hospital and at Clinica Planas, associates, residents, fellows, nurses, and secretaries, and in particular, my lymphedema nurse, Patricia Martinez, and my associate, Gemma Pons, for their constant concern and care for all our lymphedema patients. I want to take this opportunity to express my appreciation to the CRC Press team. Their professionalism has been essential to the quality of this book. Sue Hodgson deserves special mention, for without her experience and tenacity for excellence, the project would not have come together within the assigned time frame. And finally, I would like to extend a special thank you to everyone working in the field of lymphedema, for it is through the mutual sharing and exchange of knowledge that we will move forward in optimizing the quality of life for our patients. J.M.

xv

K23349_00_Neligan_FM_m6_cl_i-xxii.indd 15

5/29/2015 9:24:26 AM

xvi

Acknowledgments

A plethora of people initiated and maintained my interest and enthusiasm for all things lymphologic, but there are four in particular to whom I am particularly indebted. These are Dr. John Casley-Smith, originally from the Electron Optics Unit in the School of Medicine at the University of Adelaide, who in the late 1960s introduced me to lymphedema and who taught me all I know about the importance of the ultrastructure of the microcirculation and of the blood-tissue-lymph system; Prof. Dr. Leo Clodius from the Plastische und Wiederherstellungs Chirurgie Department of the Kantonsspital in Zurich, with whom I worked in 1975, and who first drew my attention to the range of surgical treatments that could help lymphedema, taught me about surgical models for lymphedema, and who in my view was an unacknowledged early leader in microsurgical interventions for lymphedema; and Drs. Michael Földi and Etelka Földi, with whom I worked in 1977 and who made me aware of the deficits in our lymphatic and lymphedema knowledge, who led our awareness and knowledge of how conservative treatment can make a difference, and who made me think more critically about all we say and do regarding lymphedema. N.B.P.

K23349_00_Neligan_FM_m6_cl_i-xxii.indd 16

5/29/2015 9:24:26 AM

C ontents Part I  Current Concepts in Lymphedema

 1

Lymphedema: Lack of Solutions to a Clinical Problem 

 2

Lymphedema and Its Impact on Quality of Life 

 3

Quality of Life Measurement Instruments 

Manish C. Champaneria, Peter C. Neligan

3

25

Jane M. Armer, Jennifer M. Hulett, Janice N. Cormier, Bob R. Stewart, Ausanee Wanchai, Kate D. Cromwell

Vaughan Keeley

41

Part II Anatomy, Physiology, and Lymphangiogenesis

 4

Embryology 

 5

Changing Concepts in Lymphatic Pathways 

 6

Applied Anatomy 

 7

Lymphangiogenesis 

 8

Impact of Genetics on Lymphangiogenesis 

 9

Relationship Between Fat Tissue and Lymphangiogenesis 

10

Lymphatic Malformations 

11

Immune Regulation by the Peripheral Lymphatics 

53

Sandro Michelini, Miguel Amore, Lucia Tapia, Gisela Romina Pattarone, Diego Mercado, Jeremy S. Torrisi, Ira L. Savetsky, Jason C. Gardenier, Babak J. Mehrara

61

Wei-Ren Pan

97

Miguel Amore, Lucia Tapia, Gisela Romina Pattarone, Diego Mercado

113

Jeremy S. Torrisi, Ira L. Savetsky, Jason C. Gardenier, Babak J. Mehrara

Kristiana Gordon, Pia Ostergaard

121

Mauro Andrade

135

143

Sandro Michelini, Alessandro Fiorentino, Marco Cardone

David G. Hancock

163

xvii

K23349_00_Neligan_FM_m6_cl_i-xxii.indd 17

5/29/2015 9:24:26 AM

xviii

Contents

Part III  Pathophysiology and Clinical Presentation

12

Pathophysiology of Primary Lymphedema 

13

Pathophysiology of Secondary Lymphedema 

14

Dermatologic Implications of Secondary Lymphedema of the Lower Leg 

15

Filaria 

16

Lymphedema in Pediatric Patients 

17

Combined Lymphatic and Venous Failure: Phlebolymphedema 

18

Lymphedema Risk Factors in Breast Cancer 

19

Sentinel Lymph Node Biopsy Outcomes 

177

Byung-Boong Lee, James Laredo

Etelka Földi

189

Terence Ryan, S.R. Narahari, B. Vijaya, Madhur Guruprasad Aggithaya

195

215

Gurusamy Manokaran, Rajiv Agarwal, Devisha Agarwal

Cristobal Miguel Papendieck

235

Audra A. Duncan

Swetha Kambhampati, Stanley Rockson

247

255

263

Janice N. Cormier, Kate D. Cromwell, Jane M. Armer, Merrick I. Ross

Part IV Diagnosis of Lymphedema

20 21

Causes and Classification of Lymphatic Disorders  Swetha Kambhampati, Stanley Rockson

277

Basic Approaches to the Diagnosis of Lymphedema: Clinicians’ Perspective 

Yener Demirtas, Baris Yigit

22

Biomarkers 

23

Clinical Staging of Lymphedema 

24

Measuring Methods 

25

Hydromechanics of Intercellular Fluid and Lymph 

26

Radionuclide Lymphoscintigraphy 

291

299

Kerstin Buttler, Jörg Wilting

309

Sandro Michelini, Marco Cardone, Alessandro Fiorentino

Peter C. Neligan

315

Waldemar Olszewski, Marzanna T. Zaleska

Byung-Boong Lee, James Laredo

K23349_00_Neligan_FM_m6_cl_i-xxii.indd 18

327

349

5/29/2015 9:24:26 AM

xix

Contents

27

Indocyanine Green Lymphography 

28

Magnetic Resonance Lymphangiography 

365

Mitsunaga Narushima, Takumi Yamamoto, Isao Koshima

Lee M. Mitsumori

379

Part V  Current Treatment of Lymphedema

29

Conservative Treatments for Lymphedema 

395

30

Pharmacologic Treatment of Lymphedema 

407

31

Excisional Approaches for the Treatment of Lymphedema 

Neil B. Piller

Kathleen Wang

Manish C. Champaneria, Peter C. Neligan

423



31-1  Severe Lymphedema—prepared by Peter C. Neligan

32 33

Liposuction  Håkan Brorson

437

 ultiple Lymphaticovenous Anastomoses and Multiple Lymphatic-VenousM Lymphatic Anastomoses  447 Corrado Cesare Campisi, Melissa Ryan, Corradino Campisi

34

Lymphatic Grafts 

Rüdiger G.H. Baumeister

463



34-1  Lymphatic Grafts—prepared by Rüdiger G.H. Baumeister

35

Lymphaticovenular Anastomosis  Isao Koshima, Mitsunobu Harima

473



35-1  Lymphaticovenular Anastomosis—prepared by Isao Koshima 35-2  Lymphaticovenular Anastomosis—prepared by Peter C. Neligan

36

Vascularized Lymph Node Transfer  Corinne Becker

487



36-1  Endoscopic Omentum—prepared by Peter C. Neligan 36-2  Vascularized Lymph Node Transfer—prepared by Jaume Masia

37

Reverse Lymphatic Mapping  Joseph H. Dayan, Mark L. Smith

503



37-1  Reverse Lymphatic Mapping—prepared by Joseph H. Dayan

K23349_00_Neligan_FM_m6_cl_i-xxii.indd 19

5/29/2015 9:24:26 AM

xx

Contents

38

Local Flaps and Vascularized Lymph Node Transfer 

39

Transverse Myocutaneous Gracilis With Vascularized Lymph Node Transfer 

Joshua Levine, Corinne Becker

513

Sinikka Suominen, Maija Kolehmainen



519

39-1 Transverse Myocutaneous Gracilis With Vascularized Lymph Node Transfer—

40

prepared by Sinikka Suominen

Lymphatic Microsurgical Preventing Healing Approach Concept  Francesco Boccardo

527



40-1 Lymphatic Microsurgical Preventing Healing Approach (LYMPHA) Concept—prepared by Francesco Boccardo

41

Combined Surgical Treatment for Breast Cancer–Related Lymphedema  Jaume Masia

539



41-1  Combined Surgical Treatment—prepared by Jaume Masia

42

Interventional Timing 

Peter C. Neligan, Jaume Masia

553

43

Lymphedema Complications and Their Treatment 

44

Treatment of Lymphedema in India 

45

Surgical Treatment in China 

Arin K. Greene, Reid A. Maclellan

Rajiv Agarwal, Devisha Agarwal

Ningfei Liu

559

569

573

Part VI Research and Future Directions

46

Animal Models of Lymphedema 

47

 arnessing Stem Cell–Mediated Lymphangiogenesis for the Treatment of H Lymphedema  595

583

Swapna Ghanta, Daniel A. Cuzzone, Babak J. Mehrara

Ramin Shayan, Geraldine Mitchell, Anthony Penington

48

Establishment of a Lymphedema Framework 



Credits 

Christine Moffatt, Susie Murray

Index 

K23349_00_Neligan_FM_m6_cl_i-xxii.indd 20

607

617 619

5/29/2015 9:24:26 AM

Contents

xxi

DVD



31-1  Severe Lymphedema



34-1 Lymphatic Grafts



35-1 Lymphaticovenular Anastomosis



35-2 Lymphaticovenular Anastomosis



36-1 Endoscopic Omentum



36-2 Vascularized Lymph Node Transfer



37-1 Reverse Lymphatic Mapping



39-1 Transverse Myocutaneous Gracilis With Vascularized Lymph Node Transfer



40-1 Lymphatic Microsurgical Preventing Healing Approach (LYMPHA) Concept



41-1 Combined Surgical Treatment

prepared by Peter C. Neligan

prepared by Rüdiger G.H. Baumeister

prepared by Isao Koshima

prepared by Peter C. Neligan

prepared by Peter C. Neligan

prepared by Jaume Masia

prepared by Joseph H. Dayan

prepared by Sinikka Suominen

prepared by Francesco Boccardo

K23349_00_Neligan_FM_m6_cl_i-xxii.indd 21

prepared by Jaume Masia

5/29/2015 9:24:26 AM

K23349_00_Neligan_FM_m6_cl_i-xxii.indd 22

5/29/2015 9:24:26 AM

Part I

Current Concepts in Lymphedema

K23346_Neligan_01p_Lack Solutions_r3_0001-0024.indd 1

5/26/2015 7:14:08 AM

K23346_Neligan_01p_Lack Solutions_r3_0001-0024.indd 2

5/26/2015 7:14:08 AM

C hapter 1 Lymphedema: Lack of Solutions to a Clinical Problem Manish C. Champaneria, Peter C. Neligan

K ey P oints • There is no fully satisfactory treatment for lymphedema. • Current treatment is compressive or surgical. • Decongestant therapy is the benchmark of conservative treatment.

Lym

• Surgery has traditionally been excisional. • More recent surgery includes bypass operations (lymphaticovenular anastomoses) and vascularized lymph node transfer.

Lymphedema is a progressive, chronic condition that affects a significant number of people and can have deleterious effects on the physical and psychosocial health of the patient. Lymphedema, which is especially common after surgical treatment for malignancy, has traditionally been viewed as incurable or refractory. The precise cause of lymphedema is still not completely understood, and this has led to undertreatment and misdiagnosis. Even though lymphedema may be greatly ameliorated by appropriate management, many patients receive inadequate treatment, are unaware that treatment is available, or do not know where to seek help. Several recent systematic reviews have highlighted the distinct lack of evidence for the optimal management of lymphedema.1-3 There are nonsurgical options, also known as conservative treatment, that have remained unchanged for many years despite occasionally cumbersome regimens and average results. Surgical options, which include ablative operations, liposuction, and physiologic operations, have also been used, with mixed results. Despite advances in microsurgery, there is neither consensus on surgical or nonsurgical procedures nor a standardized protocol in the treatment of the patient with lymphedema. Given the lack of consensus on one particular avenue of treatment for lymphedema, the lymphedema practitioner must have multiple intervention options available. This book will present the 3

K23346_Neligan_01p_Lack Solutions_r3_0001-0024.indd 3

5/26/2015 7:14:08 AM

4

Part I  Current Concepts in Lymphedema

latest information on the diagnosis, treatment, and management available in the field to allow the practitioner to best serve the patient. This chapter presents an overview of that information and the various solutions available to the lymphedema practitioner. The ultimate goal is to raise the profile of the condition and stimulate refined research and standardized treatment protocols to improve patient care.

Lymphedema: The Clinical Problem Lymphedema is a chronic, debilitating condition that results in the disruption of the lymphatic transport system, which leads to the accumulation of protein-rich fluid in the interstitial space because of an imbalance between interstitial fluid production and transport (usually low output failure). Outside of the United States, the most common cause of secondary lymphedema is infection with the nematode Wuchereria bancrofti. This condition is also known as filariasis (see Chapter 15 for more on filariasis). With obstructed or diseased lymphatic vessels, the accumulation of fluid gives rise to stasis of proteins and interstitial fluid. According to the Starling equation, increased protein concentration (Starling forces) results in increased colloid osmotic pressure in the tissue and a net gradient of fluid toward the interstitium. This physiologic phenomenon leads to edema, fat deposition of nontransported fat molecules, and fibrocyte activation as the result of an amplified inflammatory response. It is postulated that this triad of edema, fibrosis, and fat deposition is the supposed cause of lymphedema, although in reality this is more likely the effect than the cause. In patients with chronic lymphedema, large amounts of subcutaneous adipose tissue may form. Although incompletely understood, this adipocyte proliferation may explain why conservative treatment may not completely reduce the swelling and return the affected area to its usual dimensions (Fig. 1-1). Lymphedema manifests as soft and pitting edema early in the disease and progresses to chronic induration, overgrowth, and disfigurement later on (Fig. 1-2). Lymphedema may manifest as swelling of one or more limbs and may include the corresponding quadrant of the trunk, in addition to other areas, such as the head and neck, breast, or genitalia. It is congenital, infectious, or iatrogenic, although the exact cause is still not fully understood. Lymphedema is classified as either primary or secondary based on etiologic factors. Primary lymphedema is a congenital disease of the lymphatic system. It can present in infancy when it is known as Milroy disease. When it presents in adolescence, it is known as lymphedema praecox. Another manifestation presents when the patient is in his or her 30s and is known as lymphedema tarda. In primary lymphedema, lymphatic fluid collects in the subcutaneous tissues under the epidermis because of obstruction, malformation, or underdevelopment (hypoplasia) of various lymphatic vessels. Secondary lymphedema is an acquired disease of normal lymphatic vessels through either disruption or obstruction. At birth, about 1 in 6000 people will develop primary lymphedema. The overall prevalence of lymphedema has been estimated at 0.13% to 2%. In developed countries, the main cause of lymphedema is widely assumed to be treatment for cancer. The incidence of lymphedema after breast cancer treatment ranges from 24% to 49% after mastectomy4-8 and 4% to 28% after lumpectomy.9,10 Patients requiring more aggressive surgery and radiation have a greater risk of developing lymph-

K23346_Neligan_01p_Lack Solutions_r3_0001-0024.indd 4

5/26/2015 7:14:08 AM

5

Chapter 1  Lymphedema: Lack of Solutions to a Clinical Problem

A

Interstitial fluid

Capillary

B

Increased interstitial fluid

Lymphatic capillary

Proteins and fat molecules

Interstitial fluid

Disrupted or damaged lymphatic system

Accumulation of proteins and fat molecules

Edema

Fibrosis (fibrocyte activation)

Amplified inflammatory response

Increased subcutaneous adipose tissue (adipocyte proliferation)

FIG. 1-1  A, Under normal circumstances, proteins and fat molecules are transported with the interstitial fluid in the lymphatic flow that drains the tissues. B, When this flow is blocked, proteins and fat molecules accumulate in the interstitial fluid. This increases the colloid osmotic pressure within the interstitium, increasing the gradient of fluid toward the interstitium.

FIG. 1-2  Patient with chronic lymphedema demonstrating extreme trophic skin changes, with multiple fissures and thickening of the skin. This is elephantiasis.

edema; however, the less invasive sentinel node biopsy is associated with only a 5% to 7% incidence of upper extremity lymphedema.11 In addition to breast cancer treatment, treatment for other malignancies is also associated with lymphedema: melanoma (16%), gynecologic cancers (20%), genitourinary cancers (10%), head and neck cancers (4%), and sarcomas (30%).12 However, it appears that about one quarter to one half of affected patients have other forms of lymphedema, including primary lymphedema and lymphedema associated with poor venous function, trauma, limb dependency, or cardiac disease.13

K23346_Neligan_01p_Lack Solutions_r3_0001-0024.indd 5

5/26/2015 7:14:09 AM

6

Part I  Current Concepts in Lymphedema

TABLE 1-1  International Society of Lymphology Lymphedema Staging Stage*

Clinical Description

0

Latent or subclinical condition in which swelling is not yet evident despite impaired lymph transport with subtle changes in tissue fluid or changes in subjective symptoms. It may exist months or years before overt edema occurs.

I

Early accumulation of fluid relatively high in protein content, which subsides with limb elevation. Pitting may occur.

II

Limb elevation alone rarely reduces tissue swelling, and pitting is common.

III

Lymphostatic elephantiasis in which pitting can be absent and trophic skin changes, such as acanthosis, fat deposits, and warty overgrowths, develop.

*Other classifications/authors use arabic numerals for these stages: 0, 1, 2, 3.

The International Society of Lymphology has classified lymphedema into four stages based on certain clinical parameters14 (Table 1-1). Pain and discomfort are frequent symptoms, and increased susceptibility to cellulitis can result in frequent hospitalizations and long-term dependency on antibiotics.15 Lymphedema also causes hyperkeratosis, papillomatosis, erysipelas, lymphangitis, and the development of cutaneous tumors, such as Kaposi sarcoma, lymphoma, and even lymph­ angiosarcoma.16,17 Increased limb size can interfere with mobility and affect body image.18-20 In essence, lymphedema may produce significant physical and psychological morbidity. It is a chronic condition that presently is not curable but may be alleviated by appropriate management; if ignored, it can progress and become extremely difficult to manage.

Nonsurgical Management The treatment of lymphedema is divided into nonoperative and operative methods. With both approaches, meticulous skin hygiene and care (cleansing, low pH lotions, and emollients) are vital to the success of virtually all treatment approaches. Basic range-of-motion exercises of the extremities, limb compression, and limb elevation are also helpful. Some studies support vigorous exercises under the correct conditions. However, little substantive data exist in the form of well-designed, case-control studies that compare methodologies.14 It is clear that early treatment is optimal for the best outcome. The best practice management of lymphedema involves a multidisciplinary approach that includes the following: • Exercise and movement: to enhance lymphatic and venous flow • Swelling reduction and maintenance: to reduce limb size or volume and improve subcutaneous tissue consistency through compression and/or massage • Skin care: to optimize the condition of the skin, treat any complications caused by lymphedema, and minimize the risk of cellulitis or erysipelas • Risk reduction: to avoid factors that may exacerbate lymphedema • Pain and psychosocial management

K23346_Neligan_01p_Lack Solutions_r3_0001-0024.indd 6

5/26/2015 7:14:09 AM

Chapter 1  Lymphedema: Lack of Solutions to a Clinical Problem

7

Swelling reduction is achieved through a combination of compression and exercise with or without lymphatic massage (manual lymphatic drainage [MLD] or intermittent pneumatic compression [IPC]). The precise regimen required will be determined by the site, stage, severity, and complexity of the lymphedema and the patient’s psychosocial status. Successful management of lymphedema relies on patients playing an active role in their care.

Manual Lymphatic Drainage MLD encourages fluid away from congested areas by increasing the activity of normal lymphatics and bypassing ineffective or obliterated lymph vessels. Although there is a wealth of clinical opinion advocating the benefits of MLD, massage alone does not appear to be beneficial.21-23 Based on current evidence from 10 randomized controlled trials, there is little evidence to support the use of massage alone.24 Furthermore, if performed overly vigorously, massage may damage lymphatic vessels. Deep, heavy-handed massage should be avoided, because it may damage tissues and exacerbate edema by increasing capillary filtration. The most appropriate techniques, optimal frequency, and indications for MLD, as well as the benefits of treatment, need clarification. MLD is a specialist skill that needs regular practice to maintain competence. (MLD is discussed in detail in Chapter 29.)

Combined Physical Therapy and Compression Combined physical therapy (CPT), also known as complete or complex decongestive therapy, is also a two-stage treatment protocol. In phase I, the main goals are size reduction of the affected limb and improvement of the skin. After phase I, the patient with lymphedema proceeds to phase II, an ongoing, individualized, self-management phase to maintain the gains of phase I.25 The goals of CPT are to: • Decrease swelling26,27
 • Increase lymph drainage from the congested areas28,29
 • Reduce skin fibrosis and improve the skin’s condition30
 • Enhance the patient’s functional status31
 • Relieve discomfort and improve quality of life26,27,32-36 • Reduce the risk of cellulitis and Stewart-Treves syndrome, a rare form of angiosarcoma17,37 CPT is more labor intensive than other modalities, but its efficacy in reducing lymphedema is supported by long-standing experience.38,39 It is considered the standard of treatment for several reasons. First, CPT uses skin care, manual lymph node drainage, range-of-motion exercises, and multilayer bandage wrapping. Multilayer bandage wrapping is the mainstay of conservative therapy.40 Second, phase II maintains the results of phase I with the use of a low-stretch elastic stocking or sleeve compression. Compression alone has shown some benefit. Both multilayer inelastic stockings and controlled compression therapy (the garment’s size is tailored after edema volume changes) reduce edema volume by 31%16 and 46%,41 respectively. With CPT, randomized, controlled studies have shown a mean decrease of between 40% and 60% in edema volume.21,42,43 Noncontrolled clinical trials demonstrate results similar to those of randomized trials.27,44-47

K23346_Neligan_01p_Lack Solutions_r3_0001-0024.indd 7

5/26/2015 7:14:09 AM

8

Part I  Current Concepts in Lymphedema

Compliance is imperative for successful outcomes, and even with an actively participating patient, results vary.46 The prerequisites of successful CPT are the active participation of the patient, availability of physicians, nurses, and therapists specifically educated and experienced in this method, health insurance coverage of the cost of treatment, and industry willingness to provide highquality products. Compressive bandages, when applied incorrectly, can be harmful and should be placed by professionally trained personnel. Newer devices and garments are continuously being manufactured to reduce the bandage burden and improve compliance. However, the patient must understand that CPT is not a cure but only a risk-reduction strategy.

Thermal Therapy Although combinations of heat, skin care, and external compression have been successfully used by practitioners in Europe and Asia for thousands of patients, the role and value of thermotherapy alone without compression or MLD in the management of lymphedema remain unclear without further rigorous comparative studies.48-50

Medications Drug therapy to treat lymphedema has also been extensively studied. Diuretics have been tried, especially in the first stage of CPT. However, they are not routinely used, because they can cause fluid and electrolyte imbalance and have only a marginal benefit in reducing peripheral edema.51,52 Diuretics may also increase fibrosis because of worsening protein accumulation. Benzopyrones are not routinely used in lymphedema treatment because of poor outcomes and varied formulations and dose regimens.53 Once considered effective in the treatment of lymphedema by reducing edema fluid, softening limbs, and decreasing secondary infection, definitive conclusions about the efficacy of this medication are questionable with poor quality trials.54 Liver toxicity is linked to high dosages of this drug; therefore they are not licensed for use in the United States, United Kingdom, Australia, or France.55 Proponents claim that these drugs increase macrophage activity, encouraging the lysis of protein, which in turn reduces the formation of fibrotic tissue in the lymphedematous limb.56,57 Antimicrobials have no role in reducing lymphedema and are intended to treat cellulitis, lymphangitis, or erysipelas.40 To eradicate filaria from the bloodstream, diethylcarbamazine, albendazole, or ivermectin is recommended and may cause a variable inflammatory immune response by the host with aggravation of lymphatic blockage. There is limited evidence from rigorously designed studies on the use of natural supplements for lymphedema. American horse chestnut has been reported to reduce venous edema but not lymphedema.58 Selenium, a trace element nutrient that functions as a cofactor for a reduction of antioxidant enzymes, has been reported to improve lymphedema in head and neck cancer.59,60 Bromelain, a substance found in pineapple, has antiinflammatory, anticoagulant, enzymatic, and diuretic effects. Some have wondered if bromelain use may be beneficial for lymphedema, but it has not been studied specifically for lymphedema but rather in malignancy and other diseases.61-65

K23346_Neligan_01p_Lack Solutions_r3_0001-0024.indd 8

5/26/2015 7:14:09 AM

Chapter 1  Lymphedema: Lack of Solutions to a Clinical Problem

9

Because of the potential interactions with prescription drugs and other negative side effects, patients should check with their physician or health care provider before taking any natural supplement.

Laser Therapy Recent reports with small numbers of patients have demonstrated the efficacy of low-level laser therapy in reducing lymphedema, particularly after breast cancer.66-69 A systematic review of 41 articles showed a reduction in limb volume in patients undergoing low-level laser therapy when compared with other treatments. However, because of a lack of comparison with complex physical therapy and other treatments, definitive conclusions about low-level laser therapy cannot be purported.70 Additional studies with larger numbers of patients in diverse settings are needed to confirm these findings.

Pneumomassage IPC, also known as pneumomassage, is a two-phase program in which external gradient compression is applied by a pump, followed by elastic stockings or sleeves, to maintain edema reduction. IPC reduces edema by decreasing capillary filtration, and therefore lymph formation, rather than by accelerating lymph return. Single-chamber pumps, which were used in the past, are no longer used for lymphedema. Single-chamber pumps can cause fluid to move in both directions, even toward the edematous areas. Also, the pressure in single-chamber pumps does not stimulate lymphatic flow as sequential pumps do.71 Acceptable pumps should have appliances (pump garments) with multiple chambers and sequential pressure delivery, with the chambers compressing in a specific pattern determined individually for the patient’s diagnosis and pattern of lymphedema.72 Compliance is a concern with this approach, especially with the proximal displacement of edema and development of a fibrosclerotic ring at the root of the extremity or genitalia. Obstruction of lymph may occur, causing significant genital edema. The pumps may not be suitable for use in patients with coexisting renal failure or congestive heart failure. Patients whose lymphedema is the result of cancer treatment should ideally also be free of metastasis in the limb to prevent the risk of spreading the malignancy.73 Studies of pneumomassage are conflicting; some illustrate immediate edema reduction and long-term success, whereas others show minimal improvement with use over time.73-78 One study demonstrated considerable differences in skin or device interface pressure patterns and magnitude, which may have had an impact on therapeutic outcomes.79 This is a significant concern because superficial structures may be harmed if the pressures applied in therapy are too high.80 In general, lower pressures are considered safer, but the pressure must be individualized to the patient’s diagnosis and skin condition. IPC is not a “stand-alone” treatment.

Exercise and Elevation Exercise is a common rehabilitative intervention used to reduce lymphedema. Presently there is little evidence to indicate which types, intensities, and frequencies of exercise may be safely used in the management of lymphedema. However, specific exercise is beneficial for all patients.81

K23346_Neligan_01p_Lack Solutions_r3_0001-0024.indd 9

5/26/2015 7:14:10 AM

10

Part I  Current Concepts in Lymphedema

Exercise improves muscular strength, cardiovascular function, psychological well-being, and functional capacity. Gentle resistance exercise stimulates muscle pumps and increases lymph flow; aerobic exercise increases intraabdominal pressure, which facilitates pumping of the thoracic duct.82,83 Combinations of flexibility, resistance, and aerobic exercise may be beneficial in controlling lymphedema and should be tailored to the individual.84 Patient-appropriate exercise enables the person with lymphedema to resume activity while minimizing the risk of exacerbation of swelling.81,83-86 Physical therapy referral is required for patients who have difficulty with mobility, joint function, or joint movement. Elevation of the affected limb, ideally to just above the level of the heart, is often advised to reduce swelling. Elevation acts by maximizing venous drainage and decreasing capillary pressure and lymph production.

Diet and Weight Loss Lymphedema risk increases with obesity. Thus weight loss in overweight individuals and maintenance of optimal weight in normal-weight individuals should be integral aspects of lymphedema treatment.6,87-91 In one study, weight loss alone reduced arm volume in the lymphedema arm more than the uninvolved arm of obese women with postmastectomy lymphedema.92 However, weight loss alone does not cure lymphedema. No special diet has been proved to have therapeutic value for uncomplicated peripheral lymphedema. Restricted fluid intake has not demonstrated any benefit for peripheral lymphedema reduction. In chylous reflux syndromes (for example, intestinal lymphangiectasia), a diet as low as possible or even free of long-chain triglycerides (absorbed by intestinal lacteals) and high in shortand medium-chain triglycerides (absorbed by the portal vein) is beneficial in children. Specific vitamin supplements may be needed in very low–fat or no-fat diets.

Skin Care Skin problems are common in patients with lymphedema. Swelling may produce deep skin folds in which fungal and bacterial infections can develop, causing cellulitis or erysipelas; cracks and dry areas of the skin are entry points for bacteria and fungus.93,94 Maintenance of skin integrity, meticulous hygiene, and careful management of skin problems are important to decrease the amount of fungus or bacteria and minimize the long-term complications of skin damage. The general principles of skin care are to preserve skin barrier function through washing and the use of emollients. Ordinary soaps, which usually contain detergents and no glycerin, should be avoided because they tend to dry the skin. Natural or pH neutral soaps or soaps with glycerin should be used. The perfumes and preservatives in scented products may be irritants or allergenic. In high concentrations, mineral-based and petrolatum-based products may exacerbate dry skin conditions by occluding the skin pores and preventing the natural oils from surfacing.

K23346_Neligan_01p_Lack Solutions_r3_0001-0024.indd 10

5/26/2015 7:14:10 AM

Chapter 1  Lymphedema: Lack of Solutions to a Clinical Problem

11

Low pH moisturizers should be applied to keep the skin from drying and cracking.95 Emollients reestablish the skin’s protective lipid layer, preventing further water loss and protecting the skin from bacteria and irritants. Emollients include bath oils, soap substitutes, or moisturizers (lotions, creams, and ointments). In general, ointments contain little or no water and are better skin hydrators than creams, which are better than lotions. Emollients may damage the elastic component of compression garments, and it is advisable to avoid application immediately before donning the garment. The best method of emollient application is unknown. Some practitioners recommend applying them with strokes in the direction of hair growth (toward the feet when applying to the legs) to prevent blockage of hair follicles and folliculitis. Others recommend applying emollients by stroking toward the trunk to encourage lymph drainage. There is little substantive evidence in the literature to support particular skin care products or regimens in the treatment of lymphedema except that skin care in general is important as a risk reduction strategy.

Surgical Management Traditional teaching held that if nonsurgical treatment has been tried and unsuccessful, surgical therapy should be considered, although that paradigm may be changing. Since 1912, when the first surgical procedure for lymphedema of the scrotum was described by Charles,96 newer, more advanced options have been developed. Surgical treatment of lymphedema can be classified into three categories: liposuction, ablative surgery, and physiologic surgery.

Liposuction Liposuction involves the removal of fat in the body part affected by chronic lymphedema. It is generally performed under general anesthesia and involves the creation of many small stab incisions, followed by the insertion of suction cannulas that break up, liquefy, and suction out the fat. Liposuction has been recommended as the first surgical choice in the treatment of lymphedema. Originally used for body contouring and cosmetic surgery, liposuction has shown promising results in reducing the volume of hypertrophic adipose tissue and is also an excellent option when no lymphatic vessel is found during a shunt operation. The largest case series of liposuction in women with lymphedema of the upper extremity caused by breast cancer therapy illustrated significant improvement in the appearance and symptoms, with a mean edema reduction of 106% at 4 years’ follow-up.41,97 Liposuction has also shown promising results in lower extremity lymphedema.98 The potential risk of damaging residual lymphatic vessels, and thereby worsening the lymphedema, is of concern with liposuction, in addition to a relapse of the lymphedema if compression therapy is not continued after liposuction.99 Compression therapy after liposuction is necessary for the life of the patient. The risks of liposuction include bleeding, infection, skin loss,

K23346_Neligan_01p_Lack Solutions_r3_0001-0024.indd 11

5/26/2015 7:14:10 AM

12

Part I  Current Concepts in Lymphedema

asymmetry, abnormal sensation, and progression of lymphedema. However, with minimal complications and reduced invasiveness compared with other therapies, liposuction with compression therapy is a very viable option for patients. (For a full discussion of liposuction and its efficacy in treating lymphedema, see Chapter 32.)

Ablative Surgery Ablative operations involve the excision of subcutaneous tissue, with or without skin, from the lymphedematous body part. Several types of excisional procedures have been practiced and are still used today. These range from simple excision of the lymphedematous folds or festoons to more radical procedures, such as the Charles procedure. This procedure involves the radical excision of skin, subcutaneous tissue, and deep fascia en bloc and reconstruction with a primary or staged skin graft.96 Some surgeons prefer primary skin grafting with either the skin from the excised tissue or a nonaffected area; others favor a delayed approach to skin grafting. Both the onestage97 and two-stage98 procedures report good results in function, contour, and reduction in the incidence of secondary cellulitis. Major-General Sir Richard Henry Havelock Charles (1858-1934) was a noted physician and Sergeant Surgeon to King George V. He was also a member of the Indian Medical Service and was the medical advisor to the Secretary of State for India. While in India, Charles treated several patients with lymphedema and described its surgical treatment in the textbook, A System of Treatment. His most eponymous surgery was actually for elephantiasis of the scrotum, for which he devised a surgical garment, surgical plan, and postoperative regimen, some of which are still used today. Interestingly, Charles never performed the procedure that bears his name. He performed his technique for scrotal lymphedema, and the procedure was only later transferred to the extremities. However, the Charles procedure was cited by McIndoe and attributed to Charles. Although it is rarely performed today, there is still an occasional place for it, usually in extreme cases.100,101 The Charles procedure was refined in subsequent years. In 1927 the first surgical procedure for breast cancer–related lymphedema of the upper extremity was performed by Sistrunk,102 in which a large ellipse of skin and soft tissue, including the deep fascia, was excised along the ulnar aspect of the arm. In 1967 Thompson103,104 refined the technique by raising a hinge skin flap along the lateral aspect of the limb, excising the edematous tissue and deep fascia, and burying the skin flap over the neurovascular bundle. Both refinements created a bridge between the deep and superficial lymphatic channels, but there is no evidence to support this. Servelle described a technique in which the entire affected limb underwent a two-stage reduction (first the medial aspect and later the lateral aspect of the limb).105 This has been called total superficial lymphangiectomy and is probably a modification of the Homan procedure. This is in contrast to the Charles procedure, in which only the affected part of the limb is treated, and the cosmetic outcome is mediocre at best. These ablative operations are the simplest surgeries to perform, but they are generally no longer used because of morbidities, such as scarring, ulceration, cellulitis, lymphatic fistulas, and keloids. The main complications of this debulking procedure are infection and necrosis of the skin graft, which can lead to poor cosmetic and functional results. Nevertheless, for some extreme cases, the Charles procedure continues to be a reasonable option (Fig. 1-3).

K23346_Neligan_01p_Lack Solutions_r3_0001-0024.indd 12

5/26/2015 7:14:10 AM

Chapter 1  Lymphedema: Lack of Solutions to a Clinical Problem

A

13

B

FIG. 1-3  A, Patient with filariasis of the right lower extremity. Chronic wound on the anterior shin. B, After the Charles procedure, the patient has an excellent functional outcome.

Microsurgery Physiologic operations reconstruct the lymphatic transport system by draining lymph into the lymphatic basins or venous system. Lymphatic vessel–containing tissues, such as the greater omentum, local skin, or musculocutaneous flaps, have been used to drain excess lymph fluid. Muscle flaps have been found to restore lymphatic function in obstructive lymphedema106 and regrow lymphatic vessels in microsurgical flap reconstructions.107 The greater omental flap, with its several lymphatic vessels, has been transferred to lymphedematous extremities in breast cancer patients after mastectomy.108,109 The greater omentum was raised from the abdomen with the ipsilateral gastroepiploic vessels and transferred through a subcutaneous tunnel in the chest to the limb. The omental flap has lost popularity because of surgical morbidity and a lack of objective evidence to support any benefit. In one study on the use of an omental flap in the lower extremity, the omental flap reduced leg circumference from 50% to 75%, with excellent functional improvement in walking, daily activity, and sports.110 However, this isolated study included only four patients. Other microsurgical techniques correct the underlying lymphatic pathology. These techniques include connections between lymphatic vessels and lymphatic vessels (lymphatic-lymphatic bypass), between lymphatic vessels and veins (lymphaticovenous bypass), and between lymph nodes and veins and arteries (microvascular lymph node transfer). Lymphatic-lymphatic bypass connects an obstructed lymphatic to a healthy lymphatic with another lymphatic as an interposition graft.111 One lymphatic-lymphatic approach involved harvesting a composite graft of healthy lymphatic vessels from the medial thigh and insetting under the skin of the shoulder with several lymphatic-lymphatic anastomoses in the neck. The goal was to create several bypass routes between the upper arm and supraclavicular region by microscopically identifying lymphatic vessels at each end of the graft. In addition, an anastomosis with recipient vessels in the neck and upper arm was performed according to lymphatic vessel flow. Results from this approach revealed newly created lymph pathways and faster clearance of the radioisotope, but long donor scars and donor leg lymphedema are possible risks.112

K23346_Neligan_01p_Lack Solutions_r3_0001-0024.indd 13

5/26/2015 7:14:10 AM

14

Part I  Current Concepts in Lymphedema

FIG. 1-4  Lymphaticovenular anastomosis. Isosulfan blue dye is injected intradermally distal to the incision, which stains the lymphatics. A lymphatic channel is then anastomosed to an adjacent venule. The lymphatic vessel is on the right and the venule to which it has been anastomosed is on the left.

Another approach used a vein interposition graft between the proximal and distal lymphatic vessels to bypass the lymphedematous tissue.113 An anastomosis of the lymphatic vessels to the distal end of a vein graft and lymphatic vessels in the supraclavicular area to the other end of the graft is performed. Lymphatic-lymphatic bypass is technically demanding, time-consuming, and with such delicate, thin-walled structures, easily damaged. Lymphaticovenous bypass microsurgically connects obstructed lymphatic vessels to small, superficial veins to shunt lymph fluid into the venous system. This was first described in a rat model in 1963 by Laine and Howard114 and thereafter in dogs by Yamada,115 who then applied the technique to human patients with lymphedema. Several studies have since reported on the clinical benefit.116-120 Sedlácek121 described an end-to-side lymphaticovenous anastomosis between the saphenous vein and a lymphatic vessel. More recently, Campisi et al122 implanted several lymphatic vessels into a large vein. The lymphaticovenous anastomosis remains patent if the lymphatic pressure is higher than the venous pressure, and this is best ensured by anastomosing to low-pressure subdermal venules. Multiple small transverse incisions are made in the extremity. Subdermal venules of less than 1 mm are dissected and then anastomosed to lymphatic vessels with extra fine supermicrosurgery techniques and instruments (Fig. 1-4). Multiple anastomoses have been described, either end-to-end or end-to-side, all of which had some beneficial result.123-130 Most recently, in a study of 20 consecutive patients undergoing lymphaticovenous bypass for stage 2 or 3 lymphedema, 19 patients reported significant clinical improvement, with a mean reduction of 35% at 1 year.131 Table 1-2 outlines the published results from lymphaticovenous shunt operations. Outcomes for this type of procedure are very difficult to standardize and also to interpret, making definitive conclusions about this procedure nebulous.

K23346_Neligan_01p_Lack Solutions_r3_0001-0024.indd 14

5/26/2015 7:14:10 AM

15

Chapter 1  Lymphedema: Lack of Solutions to a Clinical Problem

TABLE 1-2  Lymphaticovenous Bypass Surgery Authors

Procedure

Fox et al128

End-to-side implantation

Gloviczki et al129

Number of Limbs

Additional Treatment

Type of Measurement

Results

Follow-up

4

Volumetry

Improved: 2 Subjectively improved: 1 No change: 1

Unknown

Pneumopump, compression

End-to-side, end-to-end anastomoses

5

Circumference

Improved: 2 No change: 3

Unknown

Pneumopump, compression

O’Brien et al118

End-to-end anastomoses

46

Circumference, volumetry

34% average reduction

13.8 yr

Elevation, massage

Filippetti et al127

End-to-side anastomoses

25

Circumference, lymphoscintigraphy

Improved: 5 No change: 8

18 mo

Unknown

Yamamoto and Sugihara123

End-to-end implantation

6

Circumference

Improved: 6

25 mo

Unknown

Koshima et al130

End-to-end anastomoses

12

Circumference

47.3% average reduction

9 yr

Elevation, compression

Campisi et al122

Lymphatic shunts

194

Volumetry, lympho­ scintigraphy

.75% improved: 73% 50%-75% improved: 24% ,25% improved: 3%

15 yr

Unknown

Chang124

End-to-end anastomoses

20

Volumetry

35% average reduction

1 yr

Unknown

Lymph node transfer is another physiologic surgical treatment of lymphedema.132,133 With lymph node transfer, a recipient bed in the lymphedematous extremity is prepared with a scar excision, followed by a flap of tissue containing superficial lymph nodes. The flap is based on an artery and vein and anastomosed to an artery and vein in the recipient bed (Fig. 1-5). Lymph nodes can typically be obtained from three donor sites: inguinal, thoracic, and cervical. In this manner, upper extremity lymphedema has been treated by transplantation of inguinal lymph nodes to the axilla or elbow region in the lymphedematous limb and even to the wrist.134,135 Lymph node transfer can also be performed simultaneously during free flap breast reconstruction, such as a deep inferior epigastric perforator flap. It is believed that the transfer of healthy lymph nodes will produce vascular endothelial growth factor C,136 which promotes lymphangiogenesis and new connections between the proximal and distal lymphatic vessels.137,138 However, there are no data showing that lymphatic vessels actually regenerate from transferred nodes. In a sheep model of the transplantation of autologous lymph nodes into a nodal excision site, there was improved lymph transport, restored lymphatic transport function, and newly formed afferent and efferent lymphatic vessels.139 Lymph node transfer may further improve lymphedema when combined with the removal of any

K23346_Neligan_01p_Lack Solutions_r3_0001-0024.indd 15

5/26/2015 7:14:10 AM

16

Part I  Current Concepts in Lymphedema

A

B

C

D

E

F

FIG. 1-5  Microvascular lymph node transfer in a breast cancer patient with lymphedema of the right upper extremity. A, Preoperative markings for the supraclavicular adipofascial flap with lymph nodes. B, Preoperative markings for node excision and exposure to recipient vessels in the right axilla. C, Dissection of the adipofascial flap with lymph nodes based on the supraclavicular artery. D, Dissection of the right axilla revealing the thoracodorsal artery as a recipient site for the flap. E, The adipofascial flap based on the supraclavicular artery. There are usually several veins to choose for outflow. F, The supraclavicular adipofascial flap with lymph nodes anastomosed to the thoracodorsal vessels in the right axilla.

scar tissue that blocks lymphatic flow,140 providing enhanced immunologic function that reduces the development of infection135 and serving as an interface between the lymphatic and venous systems without actually creating the anastomosis.141 Although physiologic operations for lymphedema are becoming more popular, the optimal technique is still under debate. There is significant discrepancy between the actual surgery and clini-

K23346_Neligan_01p_Lack Solutions_r3_0001-0024.indd 16

5/26/2015 7:14:12 AM

Chapter 1  Lymphedema: Lack of Solutions to a Clinical Problem

17

cal results. This discrepancy exists because there is a lack of standardized objective assessment for lymphedema, difficulty in choosing functional lymphatics for reconstruction, and poorly defined indications for surgery. Also, lack of knowledge regarding the cause of lymphedema limits the development of definitive treatments. More research is required—in the form of both clinical and basic science—to improve the evaluation of lymphedema, understand its cause, and refine surgical procedures.

R EFERENCES 1. Tiwari P, Coriddi M, Salani R, et al. Breast and gynecologic cancer-related extremity lymphedema: a review of diagnostic modalities and management options. World J Surg Oncol 11:237, 2013. 2. Paskett ED, Dean JA, Oliveri JM, et al. Cancer-related lymphedema risk factors, diagnosis, treatment, and impact: a review. J Clin Oncol 30:3726-3733, 2012. 3. Shah C, Arthur D, Riutta J, et al. Breast-cancer related lymphedema: a review of procedure-specific incidence rates, clinical assessment AIDS, treatment paradigms, and risk reduction. Breast J 18:357361, 2012. 4. Hinrichs CS, Watroba NL, Rezaishiraz H, et al. Lymphedema secondary to postmastectomy radiation: incidence and risk factors. Ann Surg Oncol 11:573-580, 2004. 5. Ozaslan C, Kuru B. Lymphedema after treatment of breast cancer. Am J Surg 187:69-72, 2004. 6. Petrek JA, Senie RT, Peters M, et al. Lymphedema in a cohort of breast carcinoma survivors 20 years after diagnosis. Cancer 92:1368-1377, 2001. 7. Querci della Rovere G, Ahmad I, Singh P, et al. An audit of the incidence of arm lymphoedema after prophylactic level I/II axillary dissection without division of the pectoralis minor muscle. Ann R Coll Surg Engl 85:158-161, 2003. 8. Schünemann H, Willich N. [Lymphedema after breast carcinoma. A study of 5868 cases] Dtsch Med Wochenschr 122:536-541, 1997. 9. Coen JJ, Taghian AG, Kachnic LA, et al. Risk of lymphedema after regional nodal irradiation with breast conservation therapy. Int J Radiat Oncol Biol Phys 55:1209-1215, 2003. 10. Werner RS, McCormick B, Petrek J, et al. Arm edema in conservatively managed breast cancer: obesity is a major predictive factor. Radiology 180:177-184, 1991. 11. McLaughlin SA, Wright MJ, Morris KT, et al. Prevalence of lymphedema in women with breast cancer 5 years after sentinel lymph node biopsy or axillary dissection: objective measurements. J Clin Oncol 26:5213-5219, 2008. 12. Cormier JN, Askew RL, Mungovan KS, et al. Lymphedema beyond breast cancer: a systematic review and meta-analysis of cancer-related secondary lymphedema. Cancer 116:5138-5149, 2010. 13. Moffatt CJ, Franks PJ, Doherty DC, et al. Lymphoedema: an underestimated health problem. QJM 96:731-738, 2003. 14. International Society of Lymphology. The diagnosis and treatment of peripheral lymphedema: 2013 Consensus Document of the International Society of Lymphology. Lymphology 46:1-11, 2013. 15. Carroll D, Rose K. Treatment leads to significant improvement. Effect of conservative treatment on pain in lymphoedema. Prof Nurse 8:32-33, 35-36, 1992. 16. Mortimer PS. Swollen lower limb—2: lymphoedema. BMJ 320:1527-1529, 2000. 17. Ruocco V, Schwartz RA, Ruocco E. Lymphedema: an immunologically vulnerable site for development of neoplasms. J Am Acad Dermatol 47:124-127, 2002. 18. Pereira de Godoy JM, Braile DM, de Fatima Godoy M, et al. Quality of life and peripheral lymphedema. Lymphology 35:72-75, 2002. 19. Tobin MB, Lacey HJ, Meyer L, et al. The psychological morbidity of breast cancer-related arm swelling. Psychological morbidity of lymphoedema. Cancer 72:3248-3252, 1993.

K23346_Neligan_01p_Lack Solutions_r3_0001-0024.indd 17

5/26/2015 7:14:12 AM

18

Part I  Current Concepts in Lymphedema

20. Williams AF, Moffatt CJ, Franks PJ. A phenomenological study of the lived experiences of people with lymphoedema. Int J Palliat Nurs 10:279-286, 2004. 21. McNeely ML, Magee DJ, Lees AW, et al. The addition of manual lymph drainage to compression therapy for breast cancer related lymphedema: a randomized controlled trial. Breast Cancer Res Treat 86:95-106, 2004. 22. Williams AF, Vadgama A, Franks PJ, et al. A randomized controlled crossover study of manual lymphatic drainage therapy in women with breast cancer-related lymphoedema. Eur J Cancer Care (Engl) 11:254-261, 2002. 23. Badger C, Preston N, Seers K, et al. Physical therapies for reducing and controlling lymphoedema of the limbs. Cochrane Database Syst Rev Oct 18;(4):CD003141, 2004. 24. Huang TW, Tseng SH, Lin CC, et al. Effects of manual lymphatic drainage on breast cancer-related lymphedema: a systematic review and meta-analysis of randomized controlled trials. World J Surg Oncol 11:15, 2013. 25. Johnstone PA, Hawkins K, Hood S. Role of patient adherence in maintenance of results after manipulative therapy for lymphedema. J Soc Integr Oncol 4:125-129, 2006. 26. Hamner JB, Fleming MD. Lymphedema therapy reduces the volume of edema and pain in patients with breast cancer. Ann Surg Oncol 14:1904-1908, 2007. 27. Mondry TE, Riffenburgh RH, Johnstone PA. Prospective trial of complete decongestive therapy for upper extremity lymphedema after breast cancer therapy. Cancer J 10:42-48; discussion 17-19, 2004. 28. Ferrandez JC, Laroche JP, Serin D, et al. [Lymphoscintigraphic aspects of the effects of manual lymphatic drainage] J Mal Vasc 21:283-289, 1996. 29. Franzeck UK, Spiegel I, Fischer M, et al. Combined physical therapy for lymphedema evaluated by fluorescence microlymphography and lymph capillary pressure measurements. J Vasc Res 34:306-311, 1997. 30. Foldi E, Junger M, Partsch H. The science of lymphoedema bandaging. In Calne S, ed. European Wound Management Association (EWMA) Focus Document. Lymphoedema Bandaging in Practice. London: MEP Ltd, 2005. 31. Hayes SC, Janda M, Cornish B, et al. Lymphedema after breast cancer: incidence, risk factors, and effect on upper body function. J Clin Oncol 26:3536-3542, 2008. 32. Ahmed RL, Prizment A, Lazovich D, et al. Lymphedema and quality of life in breast cancer survivors: the Iowa Women’s Health Study. J Clin Oncol 26:5689-5696, 2008. 33. Cormier JN, Xing Y, Zaniletti I, et al. Minimal limb volume change has a significant impact on breast cancer survivors. Lymphology 42:161-178, 2009. 34. Hormes JM, Bryan C, Lytle LA, et al. Impact of lymphedema and arm symptoms on quality of life in breast cancer survivors. Lymphology 43:1-13, 2010. 35. Kim SJ, Yi CH, Kwon OY. Effect of complex decongestive therapy on edema and the quality of life in breast cancer patients with unilateral lymphedema. Lymphology 40:143-151, 2007. 36. Weiss JM, Spray BJ. The effect of complete decongestive therapy on the quality of life of patients with peripheral lymphedema. Lymphology 35:46-58, 2002. 37. Cooper CL, Danieletto S, Fong E, et al. Cutaneous metastasis of epithelioid haemangioendothelioma. Pathology 41:585-587, 2009. 38. Vodder E. [Vodder’s lymph drainage. A new type of chirotherapy for esthetic prophylactic and curative purposes] Asthet Med (Berl) 14:190-191, 1965. 39. Lasinski BB, McKillip Thrift K, Squire D, et al. A systematic review of the evidence for complete decongestive therapy in the treatment of lymphedema from 2004 to 2011. PM R 4:580-601, 2012. 40. Badger CM, Peacock JL, Mortimer PS. A randomized, controlled, parallel-group clinical trial comparing multilayer bandaging followed by hosiery versus hosiery alone in the treatment of patients with lymphedema of the limb. Cancer 88:2832-2837, 2000.

K23346_Neligan_01p_Lack Solutions_r3_0001-0024.indd 18

5/26/2015 7:14:12 AM

Chapter 1  Lymphedema: Lack of Solutions to a Clinical Problem

19

41. Brorson H, Svensson H. Liposuction combined with controlled compression therapy reduces arm lymphedema more effectively than controlled compression therapy alone. Plast Reconstr Surg 102:10581067; discussion 1068, 1998. 42. Andersen L, Højris I, Erlandsen M, et al. Treatment of breast-cancer-related lymphedema with or without manual lymphatic drainage—a randomized study. Acta Oncol 39:399-405, 2000. 43. Didem K, Ufuk YS, Serdar S, et al. The comparison of two different physiotherapy methods in treatment of lymphedema after breast surgery. Breast Cancer Res Treat 93:49-54, 2005. 44. Daane S, Poltoratszy P, Rockwell WB. Postmastectomy lymphedema management: evolution of the complex decongestive therapy technique. Ann Plast Surg 40:128-134, 1998. 45. Hinrichs CS, Gibbs JF, Driscoll D, et al. The effectiveness of complete decongestive physiotherapy for the treatment of lymphedema following groin dissection for melanoma. J Surg Oncol 85:187-192, 2004. 46. Ko DS, Lerner R, Klose G, et al. Effective treatment of lymphedema of the extremities. Arch Surg 133:452-458, 1998. 47. Szuba A, Cooke JP, Yousuf S, et al. Decongestive lymphatic therapy for patients with cancer-related or primary lymphedema. Am J Med 109:296-300, 2000. 48. Campisi C, Boccardo F, Tacchella M. Use of thermotherapy in management of lymphedema: clinical observations. Int J Angiol 8:73-75, 1999. 49. Mariana VF, de Fátima GG, Maria Pde G. The effect of mechanical lymph drainage accompanied with heat on lymphedema. J Res Med Sci 16:1448-1451, 2011. 50. Ohkuma M. Lymphedema treated by microwave and elastic dressing. Int J Dermatol 31:660-663, 1992. 51. Paskett ED, Stark N. Lymphedema: knowledge, treatment, and impact among breast cancer survivors. Breast J 6:373-378, 2000. 52. Rockson SG, Miller LT, Senie R, et al. American Cancer Society Lymphedema Workshop. Workgroup III: diagnosis and management of lymphedema. Cancer 83(12 Suppl American):2882-2885, 1998. 53. Casley-Smith JR, Morgan RG, Piller NB. Treatment of lymphedema of the arms and legs with 5,6-benzo-[alpha]-pyrone. N Engl J Med 329:1158-1163, 1993. 54. Badger C, Preston N, Seers K, et al. Benzo-pyrones for reducing and controlling lymphoedema of the limbs. Cochrane Database Syst Rev (2):CD003140, 2004. 55. Loprinzi CL, Sloan J, Kugler J. Coumarin-induced hepatotoxicity. J Clin Oncol 15:3167-3168, 1997. 56. Casley-Smith JR, Casley-Smith JR. The pathophysiology of lymphedema and the action of benzopyrones in reducing it. Lymphology 21:190-194, 1988. 57. Tiwari A, Cheng KS, Button M, et al. Differential diagnosis, investigation, and current treatment of lower limb lymphedema. Arch Surg 138:152-161, 2003. 58. Siebert U, Brach M, Sroczynski G, et al. Efficacy, routine effectiveness, and safety of horsechestnut seed extract in the treatment of chronic venous insufficiency. A meta-analysis of randomized controlled trials and large observational studies. Int Angiol 21:305-315, 2002. 59. Bruns F, Büntzel J, Mücke R, et al. Selenium in the treatment of head and neck lymphedema. Med Princ Pract 13:185-190, 2004. 60. Micke O, Bruns F, Mücke R, et al. Selenium in the treatment of radiation-associated secondary lymphedema. Int J Radiat Oncol Biol Phys 56:40-49, 2003. 61. Chobotova K, Vernallis AB, Majid FA. Bromelain’s activity and potential as an anti-cancer agent: current evidence and perspectives. Cancer Lett 290:148-156, 2010. 62. Cirelli MG. Treatment of inflammation and edema with bromelain. A plant proteolytic enzyme concentrate. Del Med J 34:159-167, 1962. 63. Ley CM, Tsiami A, Ni Q, et al. A review of the use of bromelain in cardiovascular diseases. Zhong Xi Yi Jie He Xue Bao 9:702-710, 2011. 64. Schafer AI, Adelman B. Plasmin inhibition of platelet function and of arachidonic acid metabolism. J Clin Invest 75:456-461, 1985.

K23346_Neligan_01p_Lack Solutions_r3_0001-0024.indd 19

5/26/2015 7:14:12 AM

20

Part I  Current Concepts in Lymphedema

65. Seligman B. Oral bromelains as adjuncts in the treatment of acute thrombophlebitis. Angiology 20:2226, 1969. 66. Ahmed Omar MT, Abd-El-Gayed Ebid A, El Morsy AM. Treatment of post-mastectomy lymphedema with laser therapy: double blind placebo control randomized study. J Surg Res 165:82-90, 2011. 67. Mahram M, Rajabi M. Treatment of lymphedema praecox through low level laser therapy (LLLT). J Res Med Sci 16:848-851, 2011. 68. Omar MT, Shaheen AA, Zafar H. A systematic review of the effect of low-level laser therapy in the management of breast cancer-related lymphedema. Support Care Cancer 20:2977-2984, 2012. 69. Ridner SH, Poage-Hooper E, Kanar C, et al. A pilot randomized trial evaluating low-level laser therapy as an alternative treatment to manual lymphatic drainage for breast cancer-related lymphedema. Oncol Nurs Forum 40:383-393, 2013. 70. E Lima MT, E Lima JG, de Andrade MF, et al. Low-level laser therapy in secondary lymphedema after breast cancer: systematic review. Lasers Med Sci 29:1289-1295, 2014. 71. Adams KE, Rasmussen JC, Darne C, et al. Direct evidence of lymphatic function improvement after advanced pneumatic compression device treatment of lymphedema. Biomed Opt Express 1:117-125, 2010. 72. Pilch U, Wozniewski M, Szuba A. Influence of compression cycle time and number of sleeve chambers on upper extremity lymphedema volume reduction during intermittent pneumatic compression. Lymphology 42:26-35, 2009. 73. Klein MJ, Alexander MA, Wright JM, et al. Treatment of adult lower extremity lymphedema with the Wright linear pump: statistical analysis of a clinical trial. Arch Phys Med Rehabil 69(3 Pt 1):202-206, 1988. 74. Johansson K, Lie E, Ekdahl C, et al. A randomized study comparing manual lymph drainage with sequential pneumatic compression for treatment of postoperative arm lymphedema. Lymphology 31:56-64, 1998. 75. Miranda F Jr, Perez MC, Castiglioni ML, et al. Effect of sequential intermittent pneumatic compression on both leg lymphedema volume and on lymph transport as semi-quantitatively evaluated by lymphoscintigraphy. Lymphology 34:135-141, 2001. 76. Richmand DM, O’Donnell TF Jr, Zelikovski A. Sequential pneumatic compression for lymphedema. A controlled trial. Arch Surg 120:1116-1169, 1985. 77. Szuba A, Achalu R, Rockson SG. Decongestive lymphatic therapy for patients with breast carcinomaassociated lymphedema. A randomized, prospective study of a role for adjunctive intermittent pneumatic compression. Cancer 95:2260-2267, 2002. 78. Zanolla R, Monzeglio C, Balzarini A, et al. Evaluation of the results of three different methods of postmastectomy lymphedema treatment. J Surg Oncol 26:210-213, 1984. 79. Mayrovitz HN. Interface pressures produced by two different types of lymphedema therapy devices. Phys Ther 87:1379-1388, 2007. 80. Segers P, Belgrado JP, Leduc A, et al. Excessive pressure in multichambered cuffs used for sequential compression therapy. Phys Ther 82:1000-1008, 2002. 81. Schmitz KH, Ahmed RL, Troxel A, et al. Weight lifting in women with breast-cancer-related lymphedema. N Engl J Med 361:664-673, 2009. 82. Cohen SR, Payne DK, Tunkel RS. Lymphedema: strategies for management. Cancer 92(4 Suppl): 980-987, 2001. 83. Johansson K, Tibe K, Weibull A, et al. Low intensity resistance exercise for breast cancer patients with arm lymphedema with or without compression sleeve. Lymphology 38:167-180, 2005. 84. Kwan ML, Cohn JC, Armer JM, et al. Exercise in patients with lymphedema: a systematic review of the contemporary literature. J Cancer Surviv 5:320-336, 2011. 85. McKenzie DC, Kalda AL. Effect of upper extremity exercise on secondary lymphedema in breast cancer patients: a pilot study. J Clin Oncol 21:463-466, 2003.

K23346_Neligan_01p_Lack Solutions_r3_0001-0024.indd 20

5/26/2015 7:14:12 AM

Chapter 1  Lymphedema: Lack of Solutions to a Clinical Problem

21

86. Moseley AL, Piller NB, Carati CJ. The effect of gentle arm exercise and deep breathing on secondary arm lymphedema. Lymphology 38:136-145, 2005. 87. Fife C. Massive localized lymphedema, a disease unique to the morbidly obese: a case study. Ostomy Wound Manage 60:30-35, 2014. 88. Fife CE, Carter MJ. Lymphedema in the morbidly obese patient: unique challenges in a unique population. Ostomy Wound Manage 54:44-56, 2008. 89. Mahamaneerat WK, Shyu CR, Stewart BR, et al. Breast cancer treatment, BMI, post-op swelling/ lymphoedema. J Lymphoedema 3:38-44, 2008. 90. Modolin ML, Cintra W Jr, Paggiaro AO, et al. Massive localized lymphedema (MLL) in bariatric candidates. Obes Surg 16:1126-1130, 2006. 91. Soran A, D’Angelo G, Begovic M, et al. Breast cancer-related lymphedema—what are the significant predictors and how they affect the severity of lymphedema? Breast J 12:536-543, 2006. 92. Shaw C, Mortimer P, Judd PA. A randomized controlled trial of weight reduction as a treatment for breast cancer-related lymphedema. Cancer 110:1868-1874, 2007. 93. Macdonald JM. Wound healing and lymphedema: a new look at an old problem. Ostomy Wound Manage 47:52-57, 2001. 94. Mallon EC, Ryan TJ. Lymphedema and wound healing. Clin Dermatol 12:89-93, 1994. 95. Vaillant L, Gironet N. [Infectious complications of lymphedema] Rev Med Interne 23(Suppl 3):403S407S, 2002. 96. Charles RH. Elephantiasis Scroti. London: Churchill, 1912. 97. Brorson H. Liposuction in arm lymphedema treatment. Scand J Surg 92:287-295, 2003. 98. Greene AK, Slavin SA, Borud L. Treatment of lower extremity lymphedema with suction-assisted lipectomy. Plast Reconstr Surg 118:118e-121e, 2006. 99. Frick A, Hoffmann JN, Baumeister RG, et al. Liposuction technique and lymphatic lesions in lower legs: anatomic study to reduce risks. Plast Reconstr Surg 103:1868-1873; discussion 1874-1875, 1999. 100. Song R, Gao X, Li S, et al. Surgical treatment of lymphedema of the lower extremity. Clin Plast Surg 9:113-117, 1982. 101. Miller TA, Wyatt LE, Rudkin GH. Staged skin and subcutaneous excision for lymphedema: a favorable report of long-term results. Plast Reconstr Surg 10:1486-1498; discussion 1499-1501, 1998. 102. Sistrunk WE. Contribution to plastic surgery: removal of scars by stages; an open operation for extensive laceration of the anal sphincter; the Kondoleon operation for elephantiasis. Ann Surg 85:185193, 1927. 103. Thompson N. The surgical treatment of chronic lymphoedema of the extremities. Surg Clin North Am 47:445-503, 1967. 104. Thompson N. Buried dermal flap operation for chronic lymphedema of the extremities. Ten-year survey of results in 79 cases. Plast Reconstr Surg 45:541-548, 1970. 105. Servelle M. Surgical treatment of lymphedema: a report on 652 cases. Surgery 101:485-495, 1967. 106. Classen DA, Irvine L. Free muscle flap transfer as a lymphatic bridge for upper extremity lymphedema. J Reconstr Microsurg 21:93-99, 2005. 107. Slavin SA, Upton J, Kaplan WD, et al. An investigation of lymphatic function following free-tissue transfer. Plast Reconstr Surg 99:730-741; discussion 742-743, 1997. 108. Goldsmith HS. Long term evaluation of omental transposition for chronic lymphedema. Ann Surg 180:847-849, 1974. 109. Goldsmith HS, De los Santos R, Beattie EJ Jr. Relief of chronic lymphedema by omental transposition. Ann Surg 166:573-585, 1967. 110. Attash SM, Al-Sheikh MY. Omental flap for treatment of long standing lymphoedema of the lower limb: can it end the suffering? Report of four cases with review of literatures. BMJ Case Rep Feb 8;2013. 111. Springer S, Koller M, Baumeister RG, et al. Changes in quality of life of patients with lymphedema after lymphatic vessel transplantation. Lymphology 44:65-71, 2011.

K23346_Neligan_01p_Lack Solutions_r3_0001-0024.indd 21

5/26/2015 7:14:12 AM

22

Part I  Current Concepts in Lymphedema

112. Baumeister RG, Siuda S. Treatment of lymphedemas by microsurgical lymphatic grafting: what is proved? Plast Reconstr Surg 85:64-74; discussion 75-76, 1990. 113. Campisi C. Use of autologous interposition vein graft in management of lymphedema: preliminary experimental and clinical observations. Lymphology 24:71-76, 1991. 114. Laine JB, Howard JM. Experimental lymphatico-venous anastomosis. Surg Forum 14:111-112, 1963. 115. Yamada Y. Studies on lymphatic venous anastomosis in lymphedema. Nagoya J Med Sci 32:1-21, 1969. 116. Demirtas Y, Ozturk N, Yapici O, et al. Supermicrosurgical lymphaticovenular anastomosis and lymphaticovenous implantation for treatment of unilateral lower extremity lymphedema. Microsurgery 29:609-618, 2009. 117. Mihara M, Murai N, Hayashi Y, et al. Using indocyanine green fluorescent lymphography and lymphatic-venous anastomosis for cancer-related lymphedema. Ann Vasc Surg 26:278.e1-e6, 2012. 118. O’Brien BM, Mellow CG, Khazanchi RK, et al. Long-term results after microlymphaticovenous anastomoses for the treatment of obstructive lymphedema. Plast Reconstr Surg 85:562-572, 1990. 119. O’Brien BM, Shafiroff BB. Microlymphaticovenous and resectional surgery in obstructive lymphedema. World J Surg 3:3-15, 121-123, 1979. 120. O’Brien BM, Sykes P, Threlfall GN, et al. Microlymphaticovenous anastomoses for obstructive lymphedema. Plast Reconstr Surg 60:197-211, 1977. 121. Sedlácek J. Lymphovenous shunt as supplementary treatment of elephantiasis of lower limbs. Acta Chir Plast 11:157-162, 1969. 122. Campisi C, Davini D, Bellini C, et al. Lymphatic microsurgery for the treatment of lymphedema. Microsurgery 26:65-69, 2006. 123. Yamamoto Y, Sugihara T. Microsurgical lymphaticovenous implantation for the treatment of chronic lymphedema. Plast Reconstr Surg 101:157-161, 1998. 124. Chang DW. Lymphaticovenular bypass for lymphedema management in breast cancer patients: a prospective study. Plast Reconstr Surg 126:752-758, 2010. 125. Yamamoto T, Koshima I, Yoshimatsu H, et al. Simultaneous multi-site lymphaticovenular anastomoses for primary lower extremity and genital lymphoedema complicated with severe lymphorrhea. J Plast Reconstr Aesthet Surg 64:812-815, 2011. 126. Yamamoto T, Narushima M, Kikuchi K, et al. Lambda-shaped anastomosis with intravascular stenting method for safe and effective lymphaticovenular anastomosis. Plast Reconstr Surg 127:1987-1992, 2011. 127. Filippetti M, Santoro E, Graziano F, et al. Modern therapeutic approaches to postmastectomy brachial lymphedema. Microsurgery 15:604-610, 1994. 128. Fox U, Montorsi M, Romagnoli G. Microsurgical treatment of lymphedemas of the limbs. Int Surg 66:53-56, 1981. 129. Gloviczki P, Fisher J, Hollier LH, et al. Microsurgical lymphovenous anastomosis for treatment of lymphedema: a critical review. J Vasc Surg 7:647-652, 1988. 130. Koshima I, Inagawa K, Urushibara K, et al. Supermicrosurgical lymphaticovenular anastomosis for the treatment of lymphedema in the upper extremities. J Reconstr Microsurg 16:437-442, 2000. 131. Suami H, Chang DW. Overview of surgical treatments for breast cancer-related lymphedema. Plast Reconstr Surg 126:1853-1863, 2010. 132. Assouad J, Becker C, Hidden G, et al. The cutaneo-lymph node flap of the superficial circumflex artery. Surg Radiol Anat 24:87-90, 2002. 133. Becker C, Hidden G. [Transfer of free lymphatic flaps. Microsurgery and anatomical study] J Mal Vasc 13:119-122, 1988. 134. Becker C, Assouad J, Riquet M, et al. Postmastectomy lymphedema: long-term results following microsurgical lymph node transplantation. Ann Surg 243:313-315, 2006. 135. Lin CH, Ali R, Chen SC, et al. Vascularized groin lymph node transfer using the wrist as a recipient site for management of postmastectomy upper extremity lymphedema. Plast Reconstr Surg 123:12651275, 2009.

K23346_Neligan_01p_Lack Solutions_r3_0001-0024.indd 22

5/26/2015 7:14:12 AM

Chapter 1  Lymphedema: Lack of Solutions to a Clinical Problem

23

136. Saaristo AM, Niemi TS, Viitanen TP, et al. Microvascular breast reconstruction and lymph node transfer for postmastectomy lymphedema patients. Ann Surg 255:468-473, 2012. 137. Lähteenvuo M, Honkonen K, Tervala T, et al. Growth factor therapy and autologous lymph node transfer in lymphedema. Circulation 123:613-620, 2011. 138. Yan A, Avraham T, Zampell JC, et al. Mechanisms of lymphatic regeneration after tissue transfer. PLoS One 6:e17201, 2011. 139. Tobbia D, Semple J, Baker A, et al. Experimental assessment of autologous lymph node transplantation as treatment of postsurgical lymphedema. Plast Reconstr Surg 124:777-786, 2009. 140. Becker C, Vasile JV, Levine JL, et al. Microlymphatic surgery for the treatment of iatrogenic lymphedema. Clin Plast Surg 39:385-398, 2012. 141. Pegu A, Flynn JL, Reinhart TA. Afferent and efferent interfaces of lymph nodes are distinguished by expression of lymphatic endothelial markers and chemokines. Lymphat Res Biol 5:91-103, 2007.

K23346_Neligan_01p_Lack Solutions_r3_0001-0024.indd 23

5/26/2015 7:14:13 AM

K23346_Neligan_01p_Lack Solutions_r3_0001-0024.indd 24

5/26/2015 7:14:13 AM

C hapter 2 Lymphedema and Its Impact on Quality of Life Jane M. Armer, Jennifer M. Hulett, Janice N. Cormier, Bob R. Stewart, Ausanee Wanchai, Kate D. Cromwell

Ly

K ey P oints • All patients undergoing lymph node dissection for any type of solid tumor are at a lifetime risk for the development of secondary lymphedema. These solid tumors include breast cancer, melanoma, gynecologic cancer, genitourinary cancer, and head/neck cancers. • Lymphedema results in negative psychosocial outcomes, poorer physical and mental well-being, social isolation, and higher economic burden. Psychological symptom clusters include emotional distress, decreased sleep, negative self-identity and decreased self-confidence, and psychological distress. • Health care providers have long believed that posttreatment late effects of cancer treatment such as lymphedema are to be expected and tolerated; this has led to survivors’ hesitation to share lymphedema concerns with their health care providers. • As cancer detection and treatment advance, a growing number of aging cancer survivors develop posttreatment multifactorial sequelae that require a multidisciplinary approach for rehabilitation and management. • Many survivors are aware of the risk for developing lymphedema after breast cancer treatment. They are hyperaware of engaging in physical activities that may trigger lymphedema or exacerbate existing lymphedema symptoms, which results in feelings of increased frustration and limitation of daily physical activities. This in turn leads to other deleterious survivorship outcomes. • Physical impairments, functional limitations, and symptoms associated with cancer-related lymphedema may be successfully treated using a multidisciplinary approach throughout the survivorship, a period that spans from the time of cancer diagnosis through all years of life. This includes a surveillance model that advocates early assessment and referral for exercise and rehabilitative therapies. Early physiotherapy intervention may reduce the risk of developing lymphedema and of progressive lymphedema. It may also be considered cost effective, because it reduces the economic burden associated with intensive rehabilitation measures and hospitalizations for infection. • Lymphedema teaching strategies should incorporate culturally specific, spiritually based interventions to promote positive worldviews and positive spiritual experiences to enhance perceptions of quality of life. Lymphedema education should focus on the promotion of positive self-efficacy strategies that reinforce a survivor’s ability to control lymphedema through early detection and early referral for rehabilitation.

25

K23346_Neligan_02_Lymph and Impact_r2_0025-0040.indd 25

5/26/2015 7:25:57 AM

26

Part I  Current Concepts in Lymphedema

Lymphedema can arise as a consequence of the treatment of breast cancer and of many other solid tumors, such as melanoma, head and neck cancers, and gynecologic and genitourinary malignancies.1 A characteristic of all of these malignancies is that they spread to the regional lymph nodes before distant disease dissemination occurs. Treatment for many of these malignancies often includes complete dissection of the involved lymph node basin, which may include axillary, inguinofemoral, or pelvic (iliac/obturator) lymph nodes. Limited research has been dedicated to determining the incidence and impact of lymphedema among survivors of these malignancies.1 It is well established that symptoms of persistent cancer-related lymphedema result in negative psychosocial outcomes, poorer physical and mental well-being, social isolation, and higher economic burden.2-6 In addition, chronic lymphedema symptoms significantly affect the overall health-related quality of life (QOL) of older female survivors.7 As techniques for cancer detection and treatment improve, a growing population of aging cancer survivors report posttreatmentrelated difficulties that are unique and multifactorial and that therefore require a multidisciplinary approach for rehabilitation and management.8,9 Historically, health care providers have held the view that posttreatment late effects of cancer treatment such as lymphedema are expected and represent minor sequelae that are to be tolerated. Qualitative data suggest that this viewpoint has resulted in survivors’ perceptions that lymphedema symptoms should be disregarded, so these individuals therefore hesitate to share lymphedema concerns with their health care providers.10 These perceptions and interactions may be some of the reasons that few survivors receive early referrals to rehabilitation programs and consequently do not undergo baseline assessments for the early detection of physical and functional impairment.10 This chapter will explore the impact of lymphedema on QOL issues, including psychosocial outcomes, functional impairment, and economic burden experienced by cancer survivors with long-term lymphedema.

Incidence of Lymphedema In developed countries, secondary lymphedema is attributed to traumatic injury to the lymphatic channels associated with surgical interventions for the treatment of cancer.11 Determining the true prevalence and incidence rates of lymphedema is complicated, because they have been shown to vary according to the treatment received, the anatomic location affected, the methods of lymphedema assessment, the criteria applied, and the duration of follow-up.1 Moreover, methods for the global tracking and reporting of lymphedema occurrence are inconsistent, fragmented, and often disease based rather than population based.12 The diagnosis of lymphedema can be challenging given the multidisciplinary nature of treatment involving different clinicians from the time of diagnosis through the duration of long-term posttreatment follow-up care.13 Although few data are available regarding lymphedema prevalence rates in developed countries, an exception is a Brazilian study that reported a prevalence rate of 44.8% among breast cancer survivors.14 The reported incidence of lymphedema among breast cancer survivors in developed countries has been estimated as 28% in Australia, 21% in Jordan, 27% in Turkey, 30% in Great Britain, and 30% to 40% in the United States.15-22 With respect to the lifetime risk for the development of lymphedema, longitudinal data from American breast cancer survivors reveal a reported

K23346_Neligan_02_Lymph and Impact_r2_0025-0040.indd 26

5/26/2015 7:25:57 AM

Chapter 2  Lymphedema and Its Impact on Quality of Life

27

incidence that varies between 6% and 94% within 5 years after treatment, depending on the methods of measurement and the criteria applied.23,24 All patients who undergo lymph node dissection for any type of solid tumor evaluation are at a lifetime risk for lymphedema.1 Lymphedema affects survivors of various malignancies, including melanoma (16%), gynecologic cancer (20%), genitourinary cancer (10%), and head/neck cancers (4%).1 Risk factors for the development of lymphedema include poor general health, advanced breast cancer, and obesity, which are associated with breast cancer–related lymphedema.13 With respect to obesity, a body mass index (BMI) of more than 30 kg/m2 at the initiation of breast cancer treatment has been shown to be associated with a 3.6-fold increase in the likelihood of developing lymphedema at 6 months compared with survivors with BMIs of less than 30 kg/m2.13 However, weight gain after breast cancer treatment during the first 30 months of survivorship has not been shown to increase the risk of late-onset lymphedema.25 Advanced age (older than 80 years) has also been shown to be associated with a lower risk of lymphedema.7

Assessment of Quality of Life Health-related QOL is a multidimensional construct that encompasses physical, functional, emotional, and social and family well-being.26-29 Physical well-being in this context refers to symptoms related to disease (for example, pain, nausea, and fatigue) as well as side effects of treatment. The physical impact of lymphedema is very different among survivors of head/neck cancers. In this patient population, swelling of the head and neck region has significant functional implications, because it may have an impact on the patient’s ability to speak and swallow. In a study of 103 survivors of head/neck cancers, Deng et al26 reported that external and internal lymphedema also affected patients’ nutritional intake and resulted in weight loss. In addition, internal lymphedema was also linked to self-reported voice-related symptoms. Functional well-being includes an individual’s ability to perform activities of daily living such as walking, bathing, and dressing oneself in addition to societal role performance. Hull27 noted the effects of lymphedema on survivors’ daily lives, which included the following: • Difficulty sleeping because of positioning of the swollen limb • Difficulty carrying items, such as heavy pots and groceries • Challenges with many forms of exercise, even walking • Problematic fitting and comfort of clothing Overall, the physical problems associated with lymphedema affect a wide range of daily activities. Emotional well-being is a measure of coping ability, and it reflects the experience of feelings that range from enjoyment to distress, whereas social well-being reflects the quality of relationships with family and friends as well as wider social interaction.28 Health utilities are related to QOL, but they are distinctly different. Health utilities assess the value assigned by populations to specific health states with the use of standardized methods. One of the most commonly used health utilities instruments is the EuroQOL-5D, which uses preference weights from the general population and which can be used to calculate quality-adjusted life-years for cost-effectiveness analyses.30,31 In a study that examined the impact of lymphedema on utility

K23346_Neligan_02_Lymph and Impact_r2_0025-0040.indd 27

5/26/2015 7:25:57 AM

28

Part I  Current Concepts in Lymphedema

measures, elevated BMI showed a strong association with decreasing utility scores for those with higher stages (stages 2 and 3) of lymphedema.30

Quality of Life Instruments A number of instruments have been used to measure QOL among breast cancer survivors. These include the validated Psychosocial Adjustment to Illness Scale (PAIS), which was used by one study to report a low to moderate relationship between lymphedema symptoms and poor function; however, the use of the PAIS did not demonstrate a correlation between lymphedema and physical, social, vocational, or sexual function measures.32,33 The EuroQOL-5D is a validated QOL tool widely used in Europe and the United States to measure five domains: self-care, mobility, usual activities, pain/discomfort, and anxiety/depression.30 In a study of 236 patients with lymphedema, many of whom were cancer survivors, lymphedema was found to be associated with lower utility values compared with the values of the general population; adjusted utility scores were lowest in patients with lower extremity lymphedema.30 The Lymphedema Quality of Life (LYMQOL) questionnaire is a self-report tool intended for use as a standard measure of QOL outcomes among patients with upper and lower limb lymphedema in clinical settings.34 The LYMQOL was adapted from the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire-Core 30 Questions (EORTC QLQ-C30),35 and it asks 38 questions of patients with upper limb lymphedema and 40 questions of patients with lower limb lymphedema to assess four domains: mood, function, body image/appearance, and symptoms. Although the LYMQOL has demonstrated validity and reliability for all domains (Cronbach’s alpha, .0.8), the tool’s responsiveness over time continues to be evaluated.36 In a systematic review of 17 QOL instruments, Pusic et al37 reported that only one study made use of a patient-reported outcome instrument, the Upper Limb Lymphedema 27 (ULL-27), which included lymphedema-specific items.38 The ULL-27 demonstrated strong psychometric properties and content validity for the purposes of lymphedema survivorship research. A study involving the ULL-27 demonstrated improvements in QOL—specifically in the social and emotional wellbeing domains—among survivors engaged in aqua therapy for lymphedema treatment.38 Generic QOL instruments such as the Medical Outcome Survey Short-Form 36 (SF-36) are not sensitive enough to detect or measure changes related to lymphedema-associated QOL.37-39 In a systematic review of the literature pertaining to QOL among patients with lower limb lymphedema, Cernal et al40 reported that a deficit exists with regard to the availability of high-quality studies. In this review, it was noted that the majority of studies also did not use validated patientreported outcome instruments specific to assessing lymphedema-associated conditions. Current expert opinion suggests that QOL instruments are inadequate for measuring the specific psychosocial factors associated with lymphedema; specifically, they do not assess the frustration associated with lymphedema management.37,40,41 Although a number of objective tools exist, data suggest that subjective tools may be more sensitive for assessing the functional and emotional impairment associated with lymphedema.42 Therefore QOL instruments specific to lymphedema and psychosocial impairment require further research and development.37,41

K23346_Neligan_02_Lymph and Impact_r2_0025-0040.indd 28

5/26/2015 7:25:57 AM

Chapter 2  Lymphedema and Its Impact on Quality of Life

29

In a retrospective trial that assessed the effect of complete decongestive therapy for patients with melanoma and lower extremity lymphedema, patients completed a questionnaire developed by the Italian Lymphedema Association.43 This instrument addressed the patient’s general medical status as well as patient-reported QOL, and it had been validated for patients with melanoma. With the use of this questionnaire, improved QOL was noted as a result of a complete decongestive therapy regimen and an active lifestyle. Preliminary data from a prospective trial of melanoma patients used the Functional Assessment of Cancer Therapy-Melanoma (FACT-M) questionnaire44 to evaluate changes in QOL over time in patients with melanoma with or without lymphedema. In this study, lower QOL was associated with lymphedema.45 In addition, patients with lower extremity melanoma, regardless of lymphedema status, reported lower QOL after surgical treatment compared with patients with upper extremity disease. The FACT is a validated 27-item cancer-specific instrument to which tumorspecific modules can be added for many different types of cancer, including breast, bladder, cervical, endometrial, nasopharyngeal, melanoma, prostate, vulvar, and other solid and nonsolid tumors.28,46,47 A study of survivors of head and neck cancers (N 5 103), which made use of the head and neck– specific module of the FACT, reported an inverse correlation between lymphedema and functional well-being. The investigators noted that the head and neck–specific subscale was sensitive to differences in QOL among those with and without lymphedema.26 The EORTC also developed and validated a tool to assess the QOL of patients with cancer.47,48 This tool has been used to evaluate survivors of gynecologic cancers, including uterine, ovarian, cervical, and breast. In a study of 263 patients who were stratified by age and evaluated using the disease-specific modules of the EORTC QLQ, women less than 45 years old were more affected by fatigue, lymphedema, poor body image, and impaired sexuality compared with women more than 45 years old.35,49 The EORTC questionnaire was also used in a study of patients with melanoma, which found no differences in overall QOL among patients with lymphedema compared with those without lymphedema.50 In a study of 63 patients who underwent ilioinguinal lymph node dissection for the treatment of various malignancies such as melanoma, squamous cell carcinoma, and vulvar cancer, the authors reported that QOL among these patients was similar to that of the general population and that lymphedema did not have an impact on overall scores or activities of daily living.51 These findings suggest that the EORTC questionnaire with diseasespecific modules may not be sensitive enough to detect lymphedema-specific issues. A systematic review of QOL and patient-reported outcomes among patients with cancer-related lower extremity lymphedema discussed five different tools used to capture patient-reported QOL.40 Among the instruments identified were those mentioned previously: PAIS, FACT, EORTC, ULL-27, and SF-36.* The SF-36 measures physical functioning as well as mental health constructs. It was also used in a pilot study of weight lifting among patients with lower extremity lymphedema

*References 28, 32, 35, 38, 39, 50.

K23346_Neligan_02_Lymph and Impact_r2_0025-0040.indd 29

5/26/2015 7:25:57 AM

30

Part I  Current Concepts in Lymphedema

(n 5 10), but no significant differences in scores at the beginning of training compared with those obtained after the completion of training were found in this small sample.52 In another study that used the SF-36 to evaluate the effects of complex decongestive therapy on QOL among 57 survivors of gynecologic cancer, decreased limb volume was associated with statistically significant improvements in QOL.53

The Complex Effects of Lymphedema on Patients’ Well-Being Psychosocial Outcomes Lymphedema directly affects QOL through a combination of negative psychological and social factors that arise from coping with lymphedema symptoms and the complexities of lymphedema management.41 Both upper extremity and lower extremity lymphedema symptoms are associated with a psychological symptom cluster that includes emotional distress, decreased sleep, negative self-identity, decreased self-confidence, and psychological distress.4,41,54 Lymphedema in the affected hand and limb is associated with high levels of psychosocial distress, and those who have been identified as having lymphedema-associated pain have the highest levels of psychosocial distress.55 In a study that examined psychosocial distress among those with chronic lymphedema symptoms (stage 2 or 3 lymphedema) compared with those with early or transient lymphedema symptoms (stage 0 or 1 lymphedema), those with transient lymphedema reported greater levels of psychosocial distress.56 A systematic review of the psychosocial impact of lymphedema found no significant differences in emotional well-being and psychological distress among those with and without lymphedema.41 This suggests that patients accept their lymphedema over time and become better able to cope—or that the tools used are not sensitive to the differences. Qualitative findings from studies identified in the systematic review indicate that people with lymphedema feel negative social effects with respect to work, sexuality, and social engagement. Psychosocial impairment has been shown to affect breast cancer survivors with lymphedema to a greater extent than survivors without lymphedema with respect to body image, appearance, sexuality, and social isolation.41 Survivors with lymphedema experience increased feelings of frustration related to a lack of understanding from the public and a lack of consideration within the workplace, which perpetuates feelings of marginalization and social abandonment.57 Survivors’ perceptions of privacy may potentially be violated as a result of limb swelling, which is a visible indication of prior cancer treatment.58 Although studies have established that survivors of breast cancer with lymphedema rely on the social support of loved ones for physical assistance and emotional comfort, the culmination of many negative social factors may result in survivors with lymphedema limiting their social exposure, which results in further perceptions of marginalization, social abandonment, social isolation, and decreased sexuality.9,41,59

K23346_Neligan_02_Lymph and Impact_r2_0025-0040.indd 30

5/26/2015 7:25:57 AM

Chapter 2  Lymphedema and Its Impact on Quality of Life

31

Psychological Distress Recent reports note that cancer survivors experience psychological distress as early as the prediagnosis (biopsy) stage and that such distress may extend throughout the posttreatment period.60 Fear of breast cancer recurrence and of the development of posttreatment lymphedema are common causes of psychological distress during the early period of survivorship, whereas chronic symptom management and greater self-care burden contribute to ongoing psychological distress throughout survivorship.57,61,62 The emotional stress associated with cancer—particularly breast cancer—is unique with respect to recovery and rehabilitation as a result of the nonlinear and unpredictable nature of posttreatment late effects.63 A recent study reported that many survivors are aware of the risk for developing lymphedema after breast cancer treatment.59 Furthermore, survivors express a hyperawareness with regard to their decisions to engage in physical activities that may trigger lymphedema or exacerbate existing lymphedema symptoms; this in turn increases their frustration and limits their daily physical activities.64

Functional Impairment The most common sign of lymphedema is swelling of the affected limb in association with symptoms of altered limb sensation (for example, tightness, numbness, and heaviness) and limb pain; therefore many cancer survivors experience difficulty performing activities of daily living.65 Advancing age has been established as a risk factor for functional impairment in survivors who are living with lymphedema.7 With many chronic diseases, limited self-care practices have been shown to be predictive of disease progression and functional impairment.3 As functional impairment becomes chronic, lymphedema also negatively affects psychological well-being.7 Experts agree that the self-management of lymphedema symptoms after breast cancer treatment may improve survivors’ QOL.66 However, problems with treatment adherence arise in response to numerous self-care modalities that are time consuming and labor intensive but that are required for effective lymphedema symptom management.3,67 To control lymphedema symptoms, many patients must wear compression bandages and engage in ongoing bandage removal and rewrapping of the limb when participating in certain activities, particularly those that result in the wrapping getting wet, such as swimming and bathing.57 Patients may also wear compression sleeves; however, these are expensive, require hand washing, and need special care to avoid stains and other damage.68,69 Patients with lymphedema who are working have the additional burden of establishing and maintaining self-care routines with schedule constraints.70 Consequently, the burden of lymphedema self-management contributes significantly to these individuals’ overall perceptions of decreased QOL.3

Economic Impact Insurance coverage and rehabilitation costs also have a considerable impact on survivors of breast cancer with lymphedema to a greater extent than they do on survivors without lymphedema. The increased costs are primarily related to increased infections and associated rehabilitative treat-

K23346_Neligan_02_Lymph and Impact_r2_0025-0040.indd 31

5/26/2015 7:25:57 AM

32

Part I  Current Concepts in Lymphedema

ments during the first 2 years after cancer treatment is initiated.2,8 For example, Shih et al2 found that American breast cancer survivors with lymphedema were twice as likely to have additional medical complications such as seromas, lymphangitis, or cellulitis that may be associated with costly diagnostic imaging studies and more frequent hospital admissions for treatment. Over a 2-year period after the initiation of breast cancer treatment, survivors with lymphedema had greater costs in medical claims compared with survivors without lymphedema, with costs ranging from $14,877 to $23,167, depending on whether cancer costs were included.2 In another cost-analysis model, Stout et al71 reported that the cost of treating patients during the early stages of lymphedema using an innovative prospective surveillance model was $636 per year, compared with $3125 per year for patients who were managed only during the late stages of lymphedema, which is representative of the traditional treatment model. These studies suggest that early physiotherapy intervention may reduce the risk of developing lymphedema and of progressive lymphedema; this type of intervention may also be cost effective, because it reduces the economic burden associated with intensive rehabilitation measures.30,71 The economic impact of lymphedema has additional implications for patients’ personal finances beyond medical and rehabilitation costs. In a study by Moffatt et al,16 it was found that 78% of cancer survivors with lymphedema reported lost work time and that 9% experienced a negative employment outcome. For many patients with lymphedema, major factors include a lack of family, social, and professional support; insufficient health insurance; and increased financial burden associated with treatment.6 Lymphedema imposes a direct negative impact on patients’ employability as a result of insufficient financial resources, unsupportive working environments, occupations that require physical labor, dealing with employers and coworkers who lack understanding, fearing the loss of one’s job or discrimination, and the accommodation of irregular work schedules.70

Family Our understanding of how spouses and family members of survivors of breast cancer cope with cancer survivorship and the late effects of cancer treatment, including lymphedema, remains minimal.57 Lymphedema symptoms are associated with self-consciousness and feelings of unattractiveness among survivors, and survivors who wear compression sleeves report a decreased sense of sexuality and sex appeal.41,64 In a recent systematic review, individuals with lymphedema reported decreased intimacy with significant others as a result of the inclusion of lymphedema into their body images.41 Survivors of breast cancer report challenges in the area of prioritizing self-care needs, particularly with respect to lymphedema risk reduction and management as well as balancing the needs of family members.57 In addition, survivors report feelings of frustration when family members hold a negative view of their required daily time commitment for lymphedema self-care and their view of lymphedema symptoms as trivial health issues.41 These reports suggest that family dynamics are affected by the physical impairment and psychosocial issues experienced by survivors of breast cancer with lymphedema, which necessitates the renegotiation of family roles.57 Families who demonstrate resilience and view the functional impairment associated with lymphedema

K23346_Neligan_02_Lymph and Impact_r2_0025-0040.indd 32

5/26/2015 7:25:57 AM

Chapter 2  Lymphedema and Its Impact on Quality of Life

33

symptoms as manageable tend to be successful when it comes to integrating lymphedema treatment as a normal part of daily family life.57

Cultural Considerations A comparison of survivors of breast cancer with lymphedema from South Africa and the United States found that both groups reported similar experiences with regard to the impact of lymphedema on activities of daily living. Both South African and American survivors expressed dissatisfaction with the education provided that pertained to lymphedema. Among South African survivors, feelings of inadequate preparation for lymphedema symptoms was expressed, whereas their American counterparts expressed a lack of guidance with regard to the management of lymphedema.72 Unique to American survivors were four common issues related to lymphedema: the management of public curiosity; the time burden associated with limb wrapping; experiences with wearing fitted clothing; and chronic lymphedema symptoms as a reminder of having had breast cancer. Survivors from both cultures reported an awareness of the importance of self-care practices for the reduction of lymphedema risk and exacerbation. Americans were more likely to wear compression sleeves on a daily basis as well as compression bandages at night, whereas South Africans reserved sleeve use primarily for traveling and gloves for specific activities such as gardening.72 Moreover, South African and American survivors both reported relying on faith to cope with lymphedema symptoms.59,72

Spirituality and Quality of Life Positive spiritual experiences have been reported to be associated with better physical outcomes and better coping with stress among individuals with chronic diseases; negative spiritual experiences have been reported to be associated with poorer mental health outcomes.73,74 Specifically, survivors of breast cancer rely on faith and social support to decrease stress and provide inner strength to cope with symptoms of lymphedema.75 In an 84-month qualitative study, many survivors of breast cancer, including those with lymphedema, attributed having positive spiritual beliefs and experiences, increased faith, compassion, gratitude, love, hope, and feelings of connectedness to God or a higher power as essential to breast cancer recovery and coping with lymphedema.59 Furthermore, perceptions of increased QOL have been shown to be influenced by positive spiritual beliefs and a positive worldview; some survivors of breast cancer have reported experiencing a degree of spiritual transcendence.59 These perceptions are common in Judeo-Christian cultures in which a higher power is believed to be essential for recovery.76 In an attempt to examine spirituality differences among different faith traditions, researchers have examined healthy individuals from five different faiths: Buddhism, Catholicism, Islam, Protestantism, and Reform Judaism. Data have demonstrated that individuals of differing faith traditions do express differing levels of spirituality and religiosity; however, all of the faith traditions were similar with respect to congregational (social) support. In addition, no differences were found with regard to physical or mental health based on faith tradition.77

K23346_Neligan_02_Lymph and Impact_r2_0025-0040.indd 33

5/26/2015 7:25:57 AM

34

Part I  Current Concepts in Lymphedema

Interventions to Enhance Quality of Life Prevention and Education Data suggest that barriers to cancer survivors’ QOL stem from a lack of time management skills, deficits in symptom self-management knowledge, and an inability to cope with psychological distress. Therefore psychosocial interventions should be considered to address these issues.62 Individualized education programs should include information that pertains to factors that predispose individuals to the development of lymphedema, including high BMI; posttreatment complications, such as postoperative swelling, infection, and seromas; and a family history of lymphedema.9 Studies have demonstrated that exercise improves physical functioning and QOL among survivors of breast cancer.78 For example, a study of survivors with lymphedema who engaged in aqua lymphatic therapy reported greater improvements in social and emotional well-being, despite the absence of detectable limb volume changes; furthermore, survivors in the control group reported worsening QOL.38 Such findings suggest that perceptions of QOL can improve with lymphedema therapy independent of limb volume reduction.38 Furthermore, physical impairments and functional limitations associated with breast cancer treatment may be successfully treated with the use of a multidisciplinary approach throughout the survivorship period, including the incorporation of a surveillance model that advocates early assessment and referral for exercise and rehabilitative therapies.38,79,80 Lymphedema teaching strategies should also incorporate culturally specific, spiritually based interventions to promote positive worldviews and positive spiritual experiences to enhance perceptions of QOL.59

Early Intervention and Rehabilitation Intervention strategies should include planning for and assessing psychosocial problems that are commonly associated with lymphedema, including depression, poor coping, poor adherence to treatment recommendations, and social isolation.81 Symptoms of depression should be evaluated for the prompt referral of patients to mental health services; other identified psychosocial problems that are not resolved within 3 months should be referred to the appropriate specialist, such as a psychologist or social worker, for additional intervention.81 Strategies for improving QOL among cancer survivors with lymphedema should include goal setting, establishing daily care routines, regular exercise, positive self-talk, establishing support groups, and making resources for additional help available.6 Diet and exercise may be the most effective therapeutic interventions for the management of lymphedema.7 Studies have shown that reducing BMI through diet can also reduce lymphedema symptoms.82 Experts propose a prospective multidisciplinary breast cancer surveillance model that includes the following components: early and ongoing surveillance for the early identification of common physical and functional impairments; the provision of patient education to reduce lymphedema risk; prompt referral for rehabilitation and exercise interventions after physical impairments are

K23346_Neligan_02_Lymph and Impact_r2_0025-0040.indd 34

5/26/2015 7:25:57 AM

Chapter 2  Lymphedema and Its Impact on Quality of Life

35

identified; and the promotion of physical activity and weight-management strategies from diagnosis throughout the extended survivorship period.80,83,84 In a recent study, 60% of survivors were shown to have at least one posttreatment side effect that was potentially amenable to rehabilitative intervention.85 Slow and progressive resistance exercises, such as weight lifting, may reduce the onset and progression of lymphedema symptoms among cancer survivors.86,87 Aqua lymphatic therapy has been shown to be beneficial for the treatment of mild to moderate lymphedema among survivors of breast cancer.38

Integration of Complementary Alternative Medicine Practices Survivors of breast cancer tend to engage in complementary and alternative medicine (CAM) practices more than other cancer survivors do as a result of the high occurrence of side effects after treatment.88 Some survivors who were followed for 84 months reported using CAM therapies to manage these symptoms, although the success rate of symptom relief related to the use of these therapies remains largely unknown.59 Although large-scale studies of these methods are few, mindfulness-based stress reduction, acupuncture, yoga, and tai chi breathing exercises appear to be safe and may increase QOL while reducing psychological distress among survivors of breast cancer.89-91 Furthermore, preliminary studies have demonstrated a positive association between long-term yoga practice and improved QOL scores among survivors of breast cancer as well as a decrease in QOL scores among survivors who discontinue yoga practice.90 Lymphedema education should focus on the promotion of positive self-efficacy strategies that reinforce a cancer survivor’s ability to control lymphedema through early detection and early referral for rehabilitation.92 Previous research has consistently demonstrated that survivors of breast cancer with lymphedema across multiple cultural settings are open to CAM interventions.88,91

Conclusion In addition to the well-documented physical issues associated with lymphedema, there are a number of psychosocial issues that are also important to consider. Patients with lymphedema often report lower QOL, increased financial burden, and reduced social interaction. When assessing patients with lymphedema, it is crucial to evaluate all aspects of well-being. Although there are a few instruments available with lymphedema-specific items, there remains a need to develop and validate reliable and sensitive lymphedema-specific tools to assess social impact, emotional wellbeing, and functional impairment. Early studies indicate that the overall impact of lymphedema on QOL varies in accordance with the lymphedema’s anatomic location. Specifically, head and neck lymphedema is associated with a significant functional impact because it involves an impairment of the ability to swallow effectively and speak, whereas individuals with lower extremity lymphedema may be impaired in their ability to complete activities of daily living. Although the focus of lymphedema research has primarily been on lymphedema related to breast cancer, it is critical to identify the risk factors, natural histories, optimal treatment regimens, and impacts of lymphedema on survivors of cancer who had various other malignancies.

K23346_Neligan_02_Lymph and Impact_r2_0025-0040.indd 35

5/26/2015 7:25:57 AM

36

Part I  Current Concepts in Lymphedema

C linical P earls • All patients with a history of lymph node dissection should receive baseline physical and functional assessments for impairment and lifetime monitoring for signs and symptoms of secondary lymphedema. • Health care providers should be aware of the common psychological symptoms associated with lymphedema, including psychological distress, decreased sleep, negative self-identity, and decreased self-confidence. • Health care providers need to regularly inquire about signs and symptoms of lymphedema to facilitate early and timely referrals to rehabilitation programs. • Early physiotherapy intervention may reduce the risk of developing lymphedema and of progressive lymphedema. • Lymphedema education should include positive self-efficacy strategies that reinforce survivors’ ability to control lymphedema through early detection and early referral for rehabilitation. • Lymphedema teaching strategies should incorporate culturally specific, spiritually sensitive interventions to promote positive worldviews and positive spiritual experiences to enhance perceptions of QOL.

R EFERENCES 1. Cormier JN, Askew RL, Mungovan KS, et al. Lymphedema beyond breast cancer: a systematic review and meta-analysis of cancer-related secondary lymphedema. Cancer 116:5138-5149, 2010. 2. Shih YC, Xu Y, Cormier JN, et al. Incidence, treatment costs, and complications of lymphedema after breast cancer among women of working age: a 2-year follow-up study. J Clin Oncol 27:2007-2014, 2009. 3. Brown JC, Cheville AL, Tchou JC, et al. Prescription and adherence to lymphedema self-care modalities among women with breast cancer-related lymphedema. Support Care Cancer 22:135-143, 2014. 4. Cormier JN, Ross MI, Gershenwald JE, et al. Prospective assessment of the reliability, validity, and sensitivity to change of the Functional Assessment of Cancer Therapy-Melanoma questionnaire. Cancer 112:2249-2257, 2008. 5. Ridner SH. The psycho-social impact of lymphedema. Lymphat Res Biol 7:109-112, 2009. 6. Fu MR, Kang Y. Psychosocial impact of living with cancer-related lymphedema. Semin Oncol Nurs 29:50-60, 2013. 7. Clough-Gorr KM, Ganz P, Silliman RA. Older breast cancer survivors: factors associated with selfreported symptoms of persistent lymphedema over 7 years of follow-up. Breast J 16:147-155, 2010. 8. Armer JM, Stewart BR, Wanchai A, et al. Rehabilitation concepts among aging survivors living with and at risk for lymphedema: a framework for assessment, enhancing strengths, and minimizing vulnerability. Top Geriatr Rehabil 28:260-268, 2012. 9. Armer JM, Hulett JM, Bernas M, et al. Best-practice guidelines in assessment, risk reduction, management, and surveillance for post-breast cancer lymphedema. Curr Breast Cancer Rep 5:134-144, 2013. 10. Binkley JM, Harris SR, Levangie PK, et al. Patient perspectives on breast cancer treatment side effects and the prospective surveillance model for physical rehabilitation for women with breast cancer. Cancer 118(8 Suppl):2207-2216, 2012. 11. Szuba A, Rockson SG. Lymphedema: classification, diagnosis and therapy. Vasc Med 3:145-156, 1998. 12. Stout NL, Brantus P, Moffatt C. Lymphoedema management: an international intersect between developed and developing countries. Similarities, differences and challenges. Glob Public Health 7:107-123, 2012.

K23346_Neligan_02_Lymph and Impact_r2_0025-0040.indd 36

5/26/2015 7:25:57 AM

Chapter 2  Lymphedema and Its Impact on Quality of Life

37

13. Ahmed R, Schmitz K, Prizment A, et al. Risk factors for lymphedema in breast cancer survivors, the Iowa Women’s Health Study. Breast Cancer Res Treat 130:981-991, 2011. 14. Paiva DM, Rodrigues VO, Cesca MG, et al. Prevalence of lymphedema in women undergoing treatment for breast cancer in a referral center in southeastern Brazil. BMC Womens Health 13:6, 2013. 15. American Cancer Society. Breast cancer: facts & figures 2013-2014, 2013. Available at www.cancer.org/ acs/groups/content/@research/documents/document/acspc-042725.pdf. 16. Moffatt CJ, Franks PJ, Doherty DC, et al. Lymphoedema: an underestimated health problem. QJM 96:731-738, 2003. 17. Bell RJ, Robinson PJ, Barallon R, et al. Lymphedema: experience of a cohort of women with breast cancer followed for 4 years after diagnosis in Victoria, Australia. Support Care Cancer 21:2017-2024, 2013. 18. Australian Government, Australian Institute of Health and Welfare. Australian Cancer Incidence and Mortality (ACIM) books. Breast cancer. Available at www.aihw.gov.au/acim-books/. 19. Morcos BB, Al Ahmad F, Anabtawi I, et al. Lymphedema. A significant health problem for women with breast cancer in Jordan. Saudi Med J 34:62-66, 2013. 20. Norman SA, Localio AR, Potashnik SL, et al. Lymphedema in breast cancer survivors: incidence, degree, time course, treatment, and symptoms. J Clin Oncol 27:390-397, 2009. 21. Paskett ED, Naughton MJ, McCoy TP, et al. The epidemiology of arm and hand swelling in premenopausal breast cancer survivors. Cancer Epidemiol Biomarkers Prev 16:775-782, 2007. 22. Ugur S, Arici C, Yaprak M, et al. Risk factors of breast cancer-related lymphedema. Lymphat Res Biol 11:72-75, 2013. 23. Armer JM, Stewart BR. Post-breast cancer lymphedema: incidence increases from 12 to 30 to 60 months. Lymphology 43:118-127, 2010. 24. Wilke LG, McCall LM, Posther KE, et al. Surgical complications associated with sentinel lymph node biopsy: results from a prospective international cooperative group study. Ann Surg Oncol 13:491-500, 2006. 25. Ridner SH, Dietrich MS, Stewart BR, et al. Body mass index and breast cancer treatment-related lymphedema. Support Care Cancer 19:853-857, 2011. 26. Deng J, Murphy BA, Dietrich MS, et al. Impact of secondary lymphedema after head and neck cancer treatment on symptoms, functional status, and quality of life. Head Neck 35:1026-1035, 2013. 27. Hull M. Functional and psychosocial aspects of lymphedema in women treated for breast cancer. Innov Breast Cancer Care 3:117-118, 1998. 28. Cella D, Nowinski CJ. Measuring quality of life in chronic illness: the functional assessment of chronic illness therapy measurement system. Arch Phys Med Rehabil 83(12 Suppl 2):S10-S17, 2002. 29. Cella DF, Tulsky DS, Gray G, et al. The Functional Assessment of Cancer Therapy scale: development and validation of the general measure. J Clin Oncol 11:570-579, 1993. 30. Cheville AL, Almoza M, Courmier JN, et al. A prospective cohort study defining utilities using time trade-offs and the Euroqol-5D to assess the impact of cancer-related lymphedema. Cancer 116:37223731, 2010. 31. Askew RL, Swartz RJ, Xing Y, et al. Mapping FACT-melanoma quality-of-life scores to EQ-5D health utility weights. Value Health 14:900-906, 2011. 32. Derogatis LR, Derogatis MF. The Psychosocial Adjustment to Illness Scale 21 (PAIS-SR) Administration, Scoring and Procedures Manual. Towson, MD: Clinical 22 Psychometric Research Inc, 1990. 33. Shigaki CL, Madsen R, Wanchai A, et al. Upper extremity lymphedema: presence and effect on functioning five years after breast cancer treatment. Rehabil Psychol 58:342-349, 2013. 34. Keeley VL, Veigas D, Crooks S, et al. The development of a condition-specific quality of life measure for lymphoedema (LYMQOL). Eur J Lymphol 12:36, 2004. 35. Aaronson NK, Ahmedzai S, Bergman B, et al. The European Organization for Research and Treatment of Cancer QLQ-C30: a quality-of-life instrument for use in international clinical trials in oncology. J Natl Cancer Inst 85:365-376, 1993.

K23346_Neligan_02_Lymph and Impact_r2_0025-0040.indd 37

5/26/2015 7:25:57 AM

38

Part I  Current Concepts in Lymphedema

36. Keeley V, Crooks S, Locke J, et al. A quality of life measure for limb lymphoedema (LYMQOL). J Lymphoedema 5:26-37, 2010. 37. Pusic AL, Cemal Y, Albornoz C, et al. Quality of life among breast cancer patients with lymphedema: a systematic review of patient-reported outcome instruments and outcomes. J Cancer Surviv 7:83-92, 2013. 38. Tidhar D, Katz-Leurer M. Aqua lymphatic therapy in women who suffer from breast cancer treatmentrelated lymphedema: a randomized controlled study. Support Care Cancer 18:383-392, 2010. 39. McHorney CA, Ware JE Jr, Raczek AE. The MOS 36-Item Short-Form Health Survey (SF-36): II. Psychometric and clinical tests of validity in measuring physical and mental health constructs. Med Care 31:247-263, 1993. 40. Cernal Y, Jewell S, Albornoz CR, et al. Systematic review of quality of life and patient reported outcomes in patients with oncologic related lower extremity lymphedema. Lymphat Res Biol 11:14-19, 2013. 41. Fu MR, Ridner SH, Hu SH, et al. Psychosocial impact of lymphedema: a systematic review of literature from 2004 to 2011. Psychooncology 22:1466-1484, 2013. 42. Bulley C, Gaal S, Coutts F, et al. Comparison of breast cancer-related lymphedema (upper limb swelling) prevalence estimated using objective and subjective criteria and relationship with quality of life. Biomed Res Int 2013:807569, 2013. 43. Carmeli E, Bartoletti R. Retrospective trial of complete decongestive physical therapy for lower extremity secondary lymphedema in melanoma patients. Support Care Cancer 19:141-147, 2011. 44. Cormier JN, Davidson L, Xing Y, et al. Measuring quality of life in patients with melanoma: development of the FACT-Melanoma subscale. J Support Oncol 3:139-145, 2005. 45. Hyngstrom JR, Chiang YJ, Cromwell KD, et al. Prospective assessment of lymphedema incidence and lymphedema-associated symptoms following lymph node surgery for melanoma. Melanoma Res 23:290-297, 2013. 46. Webster K, Cella D, Yost K. The Functional Assessment of Chronic Illness Therapy (FACIT) Measurement System: properties, applications, and interpretation. Health Qual Life Outcomes 1:79, 2003. 47. Fayers P, Bottomley A; EORTC Quality of Life Group; Quality of Life Unit. Quality of life research within the EORTC–the EORTC QLQ-C30. European Organisation for Research and Treatment of Cancer. Eur J Cancer 38(Suppl 4):S125-S133, 2002. 48. Groenvold M, Klee MC, Sprangers MA, et al. Validation of the EORTC QLQ-C30 quality of life questionnaire through combined qualitative and quantitative assessment of patient-observer agreement. J Clin Epidemiol 50:441-450, 1997. 49. Bifulco G, De Rosa N, Tornesello ML, et al. Quality of life, lifestyle behavior and employment experience: a comparison between young and midlife survivors of gynecology early stage cancers. Gynecol Oncol 124:444-451, 2012. 50. de Vries M, Hoekstra HJ, Hoekstra-Weebers JE. Quality of life after axillary or groin sentinel lymph node biopsy, with or without completion lymph node dissection, in patients with cutaneous melanoma. Ann Surg Oncol 16:2840-2847, 2009. 51. Brouns E, Donceel P, Stas M. Quality of life and disability after ilioinguinal lymphadenectomy. Acta Chir Belg 108:685-690, 2008. 52. Katz E, Dugan NL, Cohn JC, et al. Weight lifting in patients with lower-extremity lymphedema secondary to cancer: a pilot and feasibility study. Arch Phys Med Rehabil 91:1070-1076, 2010. 53. Kim SJ, Park YD. Effects of complex decongestive physiotherapy on the oedema and the quality of life of lower unilateral lymphoedema following treatment for gynecological cancer. Eur J Cancer Care (Engl) 17:463-468, 2008. 54. Dunberger G, Lindquist H, Waldenström AC, et al. Lower limb lymphedema in gynecological cancer survivors—effect on daily life functioning. Support Care Cancer 21:3063-3070, 2013. 55. Chachaj A, Malyszczak K, Pyszel K, et al. Physical and psychological impairments of women with upper limb lymphedema following breast cancer treatment. Psychooncology 19:299-305, 2010. 56. Olivieri JM, Day JM, Alfano CM, et al. Arm/hand swelling and perceived functioning among breast cancer survivors 12 years post-diagnosis: CALGB 79804. J Cancer Surviv 2:233-242, 2008.

K23346_Neligan_02_Lymph and Impact_r2_0025-0040.indd 38

5/26/2015 7:25:57 AM

Chapter 2  Lymphedema and Its Impact on Quality of Life

39

57. Radina ME, Armer JM, Stewart BR. Making self-care a priority for women at risk of breast cancerrelated lymphedema. J Fam Nurs 20:226-249, 2014. 58. Bernas M, Askew RL, Armer J, et al. Lymphedema: how do we diagnose and reduce the risk of this dreaded complication of breast cancer treatment? Curr Breast Cancer Rep 2:53-58, 2010. 59. Hulett JM, Armer JM, Stewart BR, et al. Navigating the extended survivorship continuum: breast cancer survivors’ perspectives from diagnosis through 84-months post-treatment. Presented at the Twentyfourth Congress of the International Society of Lymphology, Rome, Italy, Sept 2013. 60. Witek-Janusek L, Gabram S, Mathews HL. Psychologic stress, reduced NK cell activity, and cytokine dysregulation in women experiencing diagnostic breast biopsy. Psychoneuroendocrinology 32:22-35, 2007. 61. Disa JJ, Petrek J. Rehabilitation after treatment for cancer of the breast. In DeVita VT Jr, Hellman S, Rosenberg SA, eds. Principles and Practice of Oncology, ed 7. Philadelphia: Lippincott Williams & Wilkins, 2005. 62. Ridner SH, Dietrich MS, Kidd N. Breast cancer treatment-related lymphedema self-care: education, practices, symptoms, and quality of life. Support Care Cancer 19:631-637, 2011. 63. Battaglini C, Dennehy C, Groff D, et al. Complementary therapies in the management of cancer treatment-related symptoms: the individualized prescriptive exercise intervention approach. Medicina Sportiva 10:49-57, 2006. 64. Radina ME. Breast cancer‐related lymphedema: implications for family leisure participation. Fam Relat 58:445-459, 2009. 65. Ridner SH. Quality of life and a symptom cluster associated with breast cancer treatment-related lymphedema. Support Care Cancer 13:904-911, 2005. 66. Togawa K, Sullivan-Halley J, Ma H, et al. Symptoms of arm lymphedema and its effects on healthrelated quality of life among disease-free female breast cancer survivors. Ann Epidemiol 23:585, 2013. 67. Ridner SH, Fu MR, Wanchai A, et al. Self-management of lymphedema: a systematic review of the literature from 2004 to 2011. Nurs Res 61:291-299, 2012. 68. Casley-Smith JR, Casley-Smith JR. Modern treatment of lymphoedema. Mod Med Australia 35:70-83, 1992. 69. Rourke LL, Hunt KK, Cormier JN. Breast cancer and lymphedema: a current overview for the healthcare provider. Womens Health (Lond Engl) 6:399-406, 2010. 70. Fu MR. Cancer survivors’ views of lymphoedema management. J Lymphoedema 5:39-48, 2010. 71. Stout NL, Pfalzer LA, Springer B, et al. Breast cancer-related lymphedema: comparing direct costs of a prospective surveillance model and a traditional model of care. Phys Ther 92:152-163, 2012. 72. Wanchai A, Stewart BR, Armer JM. Experiences and management of breast cancer-related lymphoedema: a comparison between South Africa and the United States of America. Int Nurs Rev 59:117-124, 2012. 73. Johnstone B, Yoon DP. Relationships between the brief multidimensional measurement of religiousness/spirituality and health outcomes for a heterogeneous rehabilitation population. Rehabil Psychol 54:422-431, 2009. 74. Vespa A, Jacobsen PB, Spazzafumo L, et al. Evaluation of intrapsychic factors, coping styles, and spirituality of patients affected by tumors. Psychooncology 20:5-11, 2011. 75. Fu MR. Breast cancer survivors’ intentions of managing lymphedema. Cancer Nurs 28:446-457; quiz 458-459, 2005. 76. Levin J. How faith heals: a theoretical model. Explore (NY) 5:77-96, 2009. 77. Johnstone B, Yoon DP, Cohen D, et al. Relationships among spirituality, religious practices, personality factors, and health for five different faith traditions. J Relig Health 51:1017-1041, 2012. 78. McNeely ML, Peddle CJ, Yurick JL, et al. Conservative and dietary interventions for cancer-related lymphedema: a systematic review and meta-analysis. Cancer 117:1136-1148, 2011. 79. Schmitz KH, Stout NL, Andrews K, et al. Prospective evaluation of physical rehabilitation needs in breast cancer survivors: a call to action. Cancer 118(8 Suppl):2187-2190, 2012.

K23346_Neligan_02_Lymph and Impact_r2_0025-0040.indd 39

5/26/2015 7:25:57 AM

40

Part I  Current Concepts in Lymphedema

80. Stout NL, Binkley JM, Schmitz KH, et al. A prospective surveillance model for rehabilitation for women with breast cancer. Cancer 118(8 Suppl):2191-2200, 2012. 81. Lymphoedema Framework. Best practice for the management of lymphoedema. International consensus, 2006. Available at www.woundsinternational.com/pdf/content_175.pdf. 82. Shaw C, Mortimer P, Judd P. A randomized controlled trial of weight reduction as a treatment for breast cancer-related lymphedema. Cancer 110:1868-1874, 2007. 83. Gerber LH, Stout NL, Schmitz KH, et al. Integrating a prospective surveillance model for rehabilitation into breast cancer survivorship care. Cancer 118(8 Suppl):2201-2206, 2012. 84. American College of Surgeons. National accreditation program for breast centers. Available at www. facs.org/quality-programs/napbc. 85. Schmitz KH, Stout NL, Andrews K, et al. Prospective evaluation of physical rehabilitation needs in breast cancer survivors: a call to action. Cancer 118(8 Suppl):2187-2190, 2012. 86. Schmitz KH, Ahmed RL, Troxel AB, et al. Weight lifting for women at risk for breast cancer-related lymphedema: a randomized trial. JAMA 304:2699-2705, 2010. 87. Kwan ML, Cohn JC, Armer JM, et al. Exercise in patients with lymphedema: a systematic review of the contemporary literature. J Cancer Surviv 5:320-336, 2011. 88. Wanchai A, Armer JM, Stewart BR. Performance care practices in complementary and alternative medicine by Thai breast cancer survivors: an ethnonursing study. Nurs Health Sci 14:339-344, 2012. 89. de Valois BA, Young TE, Melsome E. Assessing the feasibility of using acupuncture and moxibustion to improve quality of life for cancer survivors with upper body lymphoedema. Eur J Oncol Nurs 16:301309, 2012. 90. Douglass J, Immink M, Piller N, et al. Yoga for women with breast cancer-related lymphoedema: a preliminary 6-month study. J Lymphoedema 7:30-38, 2012. 91. Matchim Y, Armer JM, Stewart BR. Effects of mindfulness-based stress reduction (MBSR) on health among breast cancer survivors. West J Nurs Res 33:996-1016, 2011. 92. Sherman KA, Koelmeyer L. Psychosocial predictors of adherence to lymphedema risk minimization guidelines among women with breast cancer. Psychooncology 22:1120-1126, 2013.

K23346_Neligan_02_Lymph and Impact_r2_0025-0040.indd 40

5/26/2015 7:25:57 AM

C hapter 3 Quality of Life Measurement Instruments Vaughan Keeley

Qu

K ey P oints • Quality of life assessment tools have emphasized the impact of lymphedema on people’s lives. • These tools have also shown the benefits of treatment. • Validated condition-specific tools for lymphedema are available. • It is recommended that these tools be used in clinical practice and research.

In many countries, lymphedema is an underrecognized condition that is not seen as a priority by health care providers. It is often believed that lymphedema is solely related to cancer treatment, that it is rare, that it causes little in the way of morbidity, that it does not reduce life expectancy, and that there is no successful treatment for it. During recent years, growing evidence has shown that these beliefs are false. There is a greater understanding of the breadth of the problem beyond its association with cancer. This understanding includes the idea that lymphedema is more common than previously thought and causes significant morbidity among a population with an increasing life expectancy. In addition, there are a growing number of treatments that may improve lymphedema symptoms. Fundamental to this improved understanding has been research into the impact of lymphedema on an individual’s quality of life (QOL) and the improvement of QOL with treatment. This chapter will look at the QOL assessment tools available for patients with lymphedema and some of the challenges associated with the use and interpretation of these tools.

Quality of Life: Definition and Measurement Tools Defining QOL can be challenging. Every individual will have his or her own view about what constitutes a “good” QOL, and this may vary over time. Cultural and social backgrounds, age, and expectations will all influence this view. However, in the context of health-related QOL, the following components are usually considered most important1: • Physical health (symptoms and signs of illness) • Physical functioning (the ability to carry out daily activities) 41

K23346_Neligan_03_QOL Measure_r4_dc_0041-0050.indd 41

5/26/2015 7:56:38 AM

42

Part I  Current Concepts in Lymphedema

• Social health (maintaining social relationships) • Social functioning (the ability to carry out social activities) • Psychological well-being (for example, the absence of psychological distress and anxiety) • Emotional well-being (for example, life satisfaction and coping skills) More information about these components and their relationship with lymphedema can be found in Chapter 2. The World Health Organization has developed an International Classification of Functioning, Disability and Health (ICF).2 This tool aims to describe and measure health and disability, and it is particularly relevant to individuals with chronic conditions such as lymphedema. A common international approach enables the comparison of the effects of lymphedema among affected individuals in different countries, and it also allows for comparison with other conditions. The impact of a condition on an individual’s QOL can be assessed in a number of ways. Qualitative methods such as structured interviews of individuals or focus groups can be used to derive common themes that affect people with particular conditions. For example, in a study of women with lower limb lymphedema after surgery for gynecologic cancers, such themes included impact on body image and mobility and shock related to the permanence and severity of the problem.3 Quantitative methods usually involve the use of a specific QOL measurement tool in the form of a questionnaire completed by a patient. Qualitative methods such as those described previously may be used in the development of such quantitative tools to ensure their validity. Quantitative methods have the advantage of producing scores that can then be combined to describe QOL in certain populations and to show changes in scores as a result of treatment. Most QOL tools have a number of response options that represent degrees of severity. These may be divided into categories by descriptive terms or scored numerically via a visual analog scale (for example, pain can be graded on a scale of “None” to “Severe” or 0 to 10). The process of the development and validation of QOL tools should ensure that they provide meaningful results, which are discussed later in this chapter. However, most QOL life tools do not incorporate a method of “weighting” individual items and therefore can be criticized for not always accurately reflecting what matters most to a particular patient. For example, for one person, the ability to function independently may be the most important factor; for another, it may be the impact of lymphedema on body image that is of most concern. Some QOL tools, such as the Schedule for the Evaluation of Individual Quality of Life (SEIQOLDW),4 have been specifically designed to allow patients to prioritize the QOL aspects that are most important to them. Patients then rate their level of satisfaction or functioning against each of the nominated areas and indicate the relative contribution of each to their overall QOL. Similarly, the Measure Yourself Medical Outcome Profile (MYMOP) is a tool that allows patients to designate their most important symptoms, which can then be scored and reassessed after treatment to measure the outcome of an intervention.5 MYMOP also incorporates measures of selfselected physical, social, and mental activities and a general feeling of well-being.

K23346_Neligan_03_QOL Measure_r4_dc_0041-0050.indd 42

5/26/2015 7:56:38 AM

Chapter 3  Quality of Life Measurement Instruments

43

Health-related QOL tools can be general (applicable to health in general and a variety of specific conditions) or condition specific (designed to measure the impact of a particular condition). Most QOL tools cover a number of domains; these are sections that encompass common themes, such as physical health, physical and social functioning, and emotional and psychological health. Usually these domains are scored separately, and then a combined score reflects overall QOL. For patients with lymphedema, general health-related QOL scores have been used in several studies, but these may not be as accurate or informative as condition-specific tools, which have been developed more recently.6

Use of Quality of Life Tools Health-related QOL tools, including those that assess the effects of lymphedema, can be used in both clinical practice and research settings. In clinical practice, a QOL tool may facilitate the initial assessment of an individual with lymphedema by demonstrating the impact of the condition on that individual. This can then aid in the planning of treatment. If repeated measures are used, they may be able to demonstrate the effect of treatment for the individual. The combining of such outcomes of treatment for individuals seen by a lymphedema service allows QOL tools to be used in a clinical audit to demonstrate service outcomes. However, it is important that these outcomes be combined with other measures such as limb volume, some understanding of the type and severity of the lymphedema or chronic edema, the comorbidities experienced by the patient, and other environmental factors so that a realistic appraisal can be made. For example, the outcomes of treating early lymphedema related to breast cancer in otherwise healthy individuals are likely to be better than those obtained when treating complex leg edema in immobile elderly patients who may have other comorbidities and who do not have access to caretakers who will help them to put on and take off compression garments. In research, QOL tools can be used to measure the impact of lymphedema in a population, and this can help to determine the health needs of that population. These tools can also be used to assess the impact of new treatments in clinical trials. In this type of setting, QOL tools are usually combined with other outcome measures (for example, a reduction in limb volume) to obtain a more detailed assessment. QOL tools can also be used to facilitate the making of economic decisions about investments in health care. For example, these tools are used by the National Institute for Clinical Excellence in the United Kingdom. The method of cost-utility analysis involves the use of measured outcomes to create a score of health that is derived from a combination of the duration of life and an index of the patient’s health state (in other words, the patient’s QOL). The EuroQOL-5D7 is a five-item score of an individual’s health state that can be expressed on a continuum from 0.0 (death) to 1.0 (perfect health). These values are called “utilities,” and they can be combined with the length of life to give a single index known as the quality-adjusted life year (QALY). For example, living 1 year with a utility of 1.0 (perfect health) would equate to 1 QALY; living 2 years with a health utility of 0.5 would also equate to 1 QALY. Cost-utility analysis involves the use of this concept but attaches a financial cost per QALY. Thus QALYs constitute a single outcome measure that can be costed, and comparisons can be made among different treatments for a particular condition

K23346_Neligan_03_QOL Measure_r4_dc_0041-0050.indd 43

5/26/2015 7:56:38 AM

44

Part I  Current Concepts in Lymphedema

and to assess the costs of treating different conditions. This enables decisions to be made about whether to invest in certain treatments.8

Quality of Life Tools: Validation Process QOL tools to be used in research and clinical practice should undergo a robust validation process. Although methodologies may vary to some extent in different publications, there are some standard themes that are usually addressed during this process: • Validity: Is the tool measuring what it was designed to measure? • Reliability: Does the tool produce similar results under different conditions? Are results repeatable? • Responsiveness: Does the tool detect significant changes after an intervention? The details of relevant methodologies can be found in the literature that describes the validation of individual tools.9-13 There are also consensus guidelines produced by the COSMIN initiative, which seeks to improve health measurement instruments.14

Quality of Life Measures Used in Lymphedema Studies The Short Form (36) Health Survey (SF-36) has been the most commonly used QOL assessment tool, and one study has suggested that it is the most appropriate tool to use for patients who have lower limb lymphedema.15 However, this study took place before the development of most of the lymphedema-specific tools, which are described later in this chapter. The SF-36 covers eight subscales: 1. Vitality 2. Physical functioning 3. Bodily pain 4. General health perceptions 5. Physical role functioning 6. Emotional role functioning 7. Social role functioning 8. Mental health A score of 0 on this scale represents the worst possible level of health, and a score of 100 represents the best possible health. Other general QOL tools that have been used in studies of lymphedema include the following: • SF-36 Medical Outcome Study-Short Form (36 items)16 • Nottingham Health Profile Part 1 (38 items)17 • The EuroQOL Group’s EuroQOL-5D (5 items)7 • The World Health Organization Disability Assessment Scale (WHODAS) based on the ICF described previously (12- and 36-item versions)18

K23346_Neligan_03_QOL Measure_r4_dc_0041-0050.indd 44

5/26/2015 7:56:38 AM

Chapter 3  Quality of Life Measurement Instruments

45

TABLE 3-1  Examples of Generic and Cancer-Related Quality of Life Tools Used in Lymphedema Studies Quality of Life Tool

Patient Group

Number

Results

SF-3620

Chronic edema (arm and leg)

228

Reduced QOL in patients with chronic edema compared with normative data

SF-3621

Breast cancer–related lymphedema

53

QOL improved as limb volume reduced with treatment

SF-36 and EuroQOL-5D15

Lower limb lymphedema

164

SF-36 was the most appropriate tool to use for patients with lymphedema

Nottingham Health Profile Part 117

Chronic edema (arm and leg)

34

Score improved after treatment

EORTC QLQ-C30, EORTC QLQ-BR23, and WHODAS2

Breast cancer­–related lymphedema and breast cancer

283

Patients with breast cancer–related lymphedema had higher overall disability

FACT-B 1 423

Breast cancer–related lymphedema

128 (64 with breast cancer–related lymphedema)

Poorer QOL found among patients with breast cancer–related lymphedema; also used the Upper Limb Lymph­edema 27 tool and the Wesley Clinic Lymphedema Scale to assess these patients

Cancer-Specific Quality of Life Tools Much research regarding lymphedema has focused specifically on lymphedema related to cancer and particularly to breast cancer. As a result, a number of cancer-specific QOL tools have been used in studies of lymphedema related to breast cancer treatment (Table 3-1). These tools can be used together to evaluate a specific aspect of treatment, as follows: • The European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire, with 30 core items (EORTC QLQ-C30), plus the breast cancer supplement, with 23 items (EORTC QLQ-BR23) (see Chapter 2) • The Functional Assessment of Cancer Therapy-Breast Cancer, with 36 items (FACT-B), plus the FACT-B 1 4, which includes an additional four items to address issues such as arm swelling19 • The Psychosocial Adjustment to Illness Scale (PAIS) (see Chapter 2)

Condition-Specific Quality of Life Tools for Lymphedema During recent years, a number of condition-specific QOL tools for patients with lymphedema have been developed (Table 3-2). Some have been fully validated, whereas others have been used without any formal published validation. Some are designed to be used with all types of lymphedema, whereas others are specific for arm, leg, or other types of lymphedema.

K23346_Neligan_03_QOL Measure_r4_dc_0041-0050.indd 45

5/26/2015 7:56:38 AM

46

Part I  Current Concepts in Lymphedema

TABLE 3-2  Condition-Specific Quality of Life Tools for Patients With Lymphedema Number of Items

Validation Published?

Language(s)

Breast cancer– related lymphedema

5

No

English

Arm lymphedema

27

2000

French and Dutch

Quality of Life Tool

Condition

Wesley Clinic Lymphedema Scale (WCLS)24 Upper Limb Lymphedema 27 (ULL-27)25

This is validated for the assessment of QOL in patients with arm lymphedema.

Unnamed26

Peripheral lymphedema

17

No

English

Freiburg Life Quality AssessmentLymphedema (FLQA-L)9

Lymphedema

92

2005

German

Lymphedema Quality of Life Study (LYMQOL)10

Lymphedema Quality of Life Inventory (LQOLI)11

This questionnaire covers both arm and leg lymphedema and has been validated in 392 patients with primary and secondary lymphedema. It covers a number of domains: physical status, everyday life, social life, emotional well-being, treatment satisfaction, and professional/household. A shortened version of this questionnaire (the FLQA-LK, with 33 items) has been used in published studies, but no specific details regarding its validation were found in the literature.27 Arm and leg lymphedema

Arm, 33; leg, 34

2010

English and Japanese

This consists of two separate questionnaires: one for patients with arm edema and one for patients with leg edema. It covers four domains: physical symptoms, functional aspects, mood, and body image. It also includes a general QOL question. Since the publication of its validation in English, it has been translated and validated in Japanese,28 and it has also been translated into other languages. It has been used in clinical settings as part of routine assessment, and it has also been used in audit and research settings as an outcome measure. There are a few published examples of its use.29,30 It has been used in 12 countries, including some within Europe, the United States, Australia, New Zealand, and Japan. Lymphedema, all types

58 (each with three subitems)

2010

Swedish and English

This tool was originally developed in Australia. It was later translated into Swedish, and its validation was published in Sweden. It covers four domains: physical, emotional, social, and practical. It also covers all types of lymphedema, not just that related to the extremities.

Lymphedema Functioning, Disability and Health Questionnaire (Lymph-ICF)12

Breast cancer– related lymphedema

29

2011

Dutch and English

Lymphedema Functioning, Disability and Health Questionnaire for Lower Limb Lymphedema (Lymph-ICF-LL)13

Lower limb lymphedema

28

2014

Dutch and English

These two tools are linked to the World Health Organization’s International Classification of Functioning, Disability and Health. They cover five domains: physical function, mental function, general tasks and household activities, mobility activities, and life domains and social life. There are separate questionnaires for breast cancer–related lymphedema and leg lymphedema. These tools were developed in Dutch then later translated into English.

K23346_Neligan_03_QOL Measure_r4_dc_0041-0050.indd 46

5/26/2015 7:56:38 AM

Chapter 3  Quality of Life Measurement Instruments

47

Issues to Consider When Using Quality of Life Tools Comorbidity Many lymphedema clinics are now seeing patients with more complex edema of mixed causes. Among elderly patients in particular there may be significant comorbidities, such as diabetes, neurologic problems, and respiratory and cardiac conditions. These can all affect QOL, and it is often quite difficult to separate the impact of edema from other factors. For example, if a patient with a neurologic condition and limb weakness develops edema in the affected limb, the limb weakness may not improve significantly even if the edema is completely resolved with treatment.

Midline Lymphedema Many of the condition-specific QOL tools have been developed for arm and leg lymphedema and have not addressed midline edema, such as that involving the genitals, the breasts, and the head and neck. Although some tools may be used in these settings (for example, the LQOLI), there are clear functional and body image issues that may differ from those related to limb lymphedema.

The Age of the Patient All of the condition-specific QOL tools described here have been validated for adults. There is no specific lymphedema QOL tool for children.

Minimum Clinically Important Difference When using QOL tools to assess the outcomes of treatment, particularly in clinical trials, the concept of a minimum clinically important difference needs to be considered. It is possible that consistent small changes in QOL after treatment may be statistically significant. However, the question arises as to whether these changes are large enough to be of clinical importance to the patient. Ways of developing minimum clinically important differences for QOL tools have been developed but have not yet been applied to the condition-specific QOL tools for lymphedema.31

Language Translation In an ideal situation, if a QOL tool is translated into another language, it should be revalidated in that language. However, this is not always easy, and it can be very time consuming. Methods do exist for rigorous translation to ensure that meaning is not altered.32 However, cultural differences may mean that some of the items used in a particular QOL tool will not be easily transferable to other languages and cultures.

Floor and Ceiling Effects If a patient starts with a higher QOL score, there may be little room for further improvement with treatment (ceiling effect). Conversely, if he or she starts with a low QOL score, treatment can result in a large change in score (floor effect). Therefore the range of pretreatment QOL scores needs to be taken into account when assessing the effect of an intervention.

K23346_Neligan_03_QOL Measure_r4_dc_0041-0050.indd 47

5/26/2015 7:56:38 AM

48

Part I  Current Concepts in Lymphedema

Length of Questionnaire and Frequency of Use A long questionnaire or a tool that needs to be used too frequently to monitor outcomes can deter patients from compliance. Consideration should be given to both of these aspects, particularly when administering a tool to elderly patients with significant comorbidities.

Conclusion During recent years, an increasing number of condition-specific QOL tools have become available for use by patients with lymphedema. It is important that these tools are used both clinically and in research studies to strengthen the evidence of the impact of lymphedema on patient QOL and the improvement of QOL with successful treatment.

C linical P earls • The following components are usually considered most important when assessing health-related QOL: physical health, physical functioning, social health, social functioning, psychological well-being, and emotional wellbeing. • Health-related QOL tools can be general (applicable to health in general and a variety of specific conditions) or condition specific (designed to measure the impact of a particular condition). • In clinical practice, a QOL tool may facilitate the initial assessment of an individual with lymphedema by demonstrating the impact of the condition on that individual. In research, QOL tools can be used to measure the impact of lymphedema in a population, and this can help to determine the health needs of that population. QOL tools can also be used to facilitate the making of economic decisions about investments in health care. • QOL tools to be used in research and clinical practice should undergo a robust validation process. • It is important that QOL tools be used both clinically and in research studies to strengthen the evidence of the impact of lymphedema on patient QOL and the improvement of QOL with successful treatment.

R EFERENCES 1. Bowling A. Research Methods in Health: Investigating Health and Health Services, ed 1. Philadelphia: Open University Press, 1997. 2. World Health Organization. International Classification of Functioning, Disability and Health (ICF), 2014. Available at www.who.int/classifications/icf/en/. 3. Ryan M, Stainton MC, Jaconelli C, et al. The experience of lower limb lymphedema for women after treatment for gynecologic cancer. Oncol Nurs Forum 30:417-423, 2003. 4. Hickey AM, Bury G, O’Boyle CA, et al. A new short form individual quality of life measure (SEIQOLDW): application in a cohort of individuals with HIV/AIDS. BMJ 313:29-33, 1996. 5. University of Bristol, Centre for Academic Primary Care. Welcome to MYMOP. Available at www. bristol.ac.uk/primaryhealthcare/resources/mymop.

K23346_Neligan_03_QOL Measure_r4_dc_0041-0050.indd 48

5/26/2015 7:56:38 AM

Chapter 3  Quality of Life Measurement Instruments

49

6. Morgan PA, Franks PJ, Moffatt CJ. Health-related quality of life with lymphoedema: a review of the literature. Int Wound J 2:47-62, 2005. 7. EuroQOL Group. EuroQOL—a new facility for the measurement of health-related quality of life. Health Policy 16:199-208, 1990. 8. Keeley V, Franks PJ. Evaluating a lymphoedema service. Lymphoedema Framework. Template for Management: Developing a Lymphoedema Service. London: MEP Ltd, 2007. 9. Augustin M, Bross F, Földi E, et al. Development, validation and clinical use of the FLQA-1, a diseasespecific quality of life questionnaire for patients with lymphedema. VASA 34:31-35, 2005. 10. Keeley V, Crooks S, Locke J, et al. Quality of life measure for limb lymphoedema (LYMQOL). J Lymphoedema 5:26-37, 2010. 11. Klernäs P, Kristjanson LJ, Johansson K. Assessment of quality of life in lymphedema patients: validity and reliability of the Swedish version of the Lymphedema Quality of Life Inventory (LQOLI). Lymphology 43:135-145, 2010. 12. Devoogdt N, Van Kampen M, Geraerts I, et al. Lymphoedema Functioning, Disability and Health questionnaire (Lymph-ICF): reliability and validity. Phys Ther 91:944-957, 2011. 13. Devoogdt N, De Groef A, Hendrickx AD, et al. Lymphedema Functioning, Disability and Health Questionnaire for Lower Limb Lymphoedema (Lymph-ICF-LL): reliability and validity. Phys Ther 94:705721, 2014. 14. COSMIN. The COSMIN checklist. Available at www.cosmin.nl/the-cosmin-checklist_8_5.html. 15. Franks PJ, Moffatt CJ, Doherty DC, et al. Assessment of health-related quality of life in patients with lymphedema of the lower limb. Wound Repair Regen 14:110-118, 2006. 16. Ware JE, Sherbourne CD. The MOS 36-item short-form health survey (SF-36) I. Conceptual framework and item selection. Med Care 30:473-483, 1992. 17. Sitzia J, Sobrido L. Measurement of health-related quality of life of patients receiving conservative treatment for limb lymphoedema using the Nottingham Health Profile. Qual Life Res 6:373-384, 1997. 18. World Health Organization. WHO Disability Assessment Schedule 2.0 (WHODAS 2.0), 2014. Available at www.who.int/classifications/icf/whodasii/en/. 19. Coster S, Poole K, Fallowfield LJ. The validation of a quality of life scale to assess the impact of arm morbidity in breast cancer patients post-operatively. Breast Cancer Res Treat 68:273-282, 2001. 20. Moffatt CJ, Franks PJ, Doherty DC, et al. Lymphoedema: an underestimated health problem. QJM 96:731-738, 2003. 21. Kim SJ, Yi CH, Kwon OY. Effect of complex decongestive therapy on edema and the quality of life in breast cancer patients with unilateral lymphedema. Lymphology 40:143-151, 2007. 22. Pyszel A, Malyszczak K, Pyszel K, et al. Disability, psychological distress and quality of life in breast cancer survivors with arm lymphedema. Lymphology 39:185-192, 2006. 23. Ridner SH. Quality of life and a symptom cluster associated with breast cancer treatment-related lymphedema. Support Care Cancer 13:904-911, 2005. 24. Mirolo BR, Bunce IH, Chapman M, et al. Psychological benefits of postmastectomy lymphedema therapy. Cancer Nurs 18:197-205, 1995. 25. Launois R, Alliott F. Quality of life scale in upper limb lymphoedema—a validation study. Lymphology 33:266-274, 2000. 26. Weiss JM, Spray BJ. The effect of complete decongestive therapy on the quality of life of patients with peripheral lymphedema. Lymphology 35:46-58, 2002. 27. Blome C, Augustin M, Heyer K, et al. Evaluation of patient-relevant outcomes of lymphedema and lip­ edema treatment: development and validation of a new benefit tool. Eur J Vasc Endovasc Surg 47:100107, 2014. 28. Takeuchi M. Development of a quality of life measure for limb lymphoedema (LYMQOL), Japanese version. Abstract from the International Lymphoedema Framework Conference, Montpellier, France, June 2012.

K23346_Neligan_03_QOL Measure_r4_dc_0041-0050.indd 49

5/26/2015 7:56:38 AM

50

Part I  Current Concepts in Lymphedema

2 9. Hardy D. Management of a patient with secondary lymphedema. Cancer Nurs Pract 11:21-26, 2012. 30. Jeffs E, Wiseman T. Randomised controlled trial to determine the benefit of daily home-based exercise in addition to self-care in the management of breast cancer-related lymphoedema: a feasibility study. Support Care Cancer 21:1013-1023, 2013. 31. Hays RD, Woolley JM. The concept of clinically meaningful difference in health-related quality of life research. How meaningful is it? Pharmacoeconomics 18:419-423, 2000. 32. Piault E, Doshi S, Brandt BA, et al. Linguistic validation of translation of the Self-Assessment Goal Achievement (SAGA) questionnaire from English. Health Qual Life Outcomes 10:40, 2012.

K23346_Neligan_03_QOL Measure_r4_dc_0041-0050.indd 50

5/26/2015 7:56:38 AM

Part II

Anatomy, Physiology, and Lymphangiogenesis

K23346_Neligan_04_Embryology_r5_0051-0060.indd 51

5/26/2015 7:58:55 AM

K23346_Neligan_04_Embryology_r5_0051-0060.indd 52

5/26/2015 7:58:55 AM

C hapter 4 Embryology Sandro Michelini, Miguel Amore, Lucia Tapia, Gisela Romina Pattarone, Diego Mercado, Jeremy S. Torrisi, Ira L. Savetsky, Jason C. Gardenier, Babak J. Mehrara

K ey P oints • The lymphatic system has several critical roles, including transporting tissue fluids and plasma protein back to the bloodstream and providing major pathways for the spread of metastatic tumor cells. • The development of the lymphatic vasculature is a stepwise process requiring the specification of lymphatic endothelial cell progenitors from the embryonic veins. • The embryology of the vascular and lymphatic systems during gestation must be considered collectively because they are interconnected.

A

Em

The lymphatic system transports tissue fluids and extravasated plasma proteins back to the bloodstream and absorbs lipids from the intestinal tract. The lymphatic system also plays a crucial role in immune response and is one of the main routes for the metastatic spread of tumor cells. The developmental origin of the lymphatic system remained unclear until the first years of the twentieth century, when Sabine proposed (after extensive ink injection experiments in pig embryos) that the jugular sacs arose from the anterior cardinal vein. Recent analyses of targeted gene deletions and genetic lineage tracing studies in mice have confirmed Sabine’s models.1

Overview of the Lymphatic System The lymphatic system is the third vascular system. A major function of the lymphatic vasculature is to transport extracellular fluid back to the cardiovascular system. When blood enters a capillary bed, high hydrostatic pressure on the arterial side of the capillary causes plasma to leak into the interstitial space. On the venous side of the capillary bed, the inward pull of the blood’s high osmotic pressure exceeds the outward push of its hydrostatic pressure, causing the previously filtered fluid 53

K23346_Neligan_04_Embryology_r5_0051-0060.indd 53

5/26/2015 7:58:56 AM

54

Part II  Anatomy, Physiology, and Lymphangiogenesis

Right jugular trunk Right lymphatic duct Right subclavian trunk Right subclavian vein Right bronchomediastinal trunk

Left jugular trunk Left subclavian trunk Left subclavian vein Left bronchomediastinal trunk Thoracic duct

Cisterna chyli Lumbar trunks

Intestinal trunk

To right lymphatic duct To thoracic duct

FIG. 4-1  The lymphatic system of the upper extremity network of capillaries and collecting lymphatics. General pattern of lymph drainage to the right lymphatic and thoracic ducts.

to rush back into the capillary bed and reenter the systemic circulation. This system allows 90% of the filtered fluid to reenter the circulation; the remaining 10% is returned to the systemic circulation by the lymphatic system through a series of successively larger lymphatic vessels. Lymph is a fluid that is similar in composition to plasma but contains leukocytes and higher concentrations of macromolecules, such as proteins and lipids. Lymph enters the lymphatic system through lymphatic capillaries that are located near blood capillaries and is subsequently transferred to larger lymphatics, which are known as collecting lymphatic vessels (Figs. 4-1 and 4-2). These collecting lymphatic vessels then transfer the lymph unidirectionally toward the chest. Collecting lymphatics from the left side of the body, the abdomen, and from both legs drain lymph into the thoracic duct, which then drains the lymph into the left subclavian vein. Lymph from the right side of the body, head, and thorax drains into the right subclavian vein after it first drains into

K23346_Neligan_04_Embryology_r5_0051-0060.indd 54

5/26/2015 7:58:56 AM

55

Chapter 4  Embryology

Lymphatic capillary plexus Skin

Epidermis Dermis

Lymphatic precollector vessel Superficial lymphatic collector vessel (afferent) Superficial lymph node

Subcutaneous tissue

Lymphatic collector vessel (efferent)

Deep fascia

Subfascial layer

To venous system and systemic circulation

Muscle

Perforating vessel

Deep lymphatic collector vessel

Deep lymph node

FIG. 4-2  Structure of the superficial and deep lymphatic system.

50 mm

FIG. 4-3  Capillary (red arrows) and collecting lymphatic (white arrow) vessels in a mouse hindlimb. Note the layer of smooth muscle cells on the collecting lymphatic vessel. (LYVE, Lymphatic vessel endothelial hyaluronan receptor; aSMA, alpha-smooth muscle actin; DAPI, 49, 6-diamidino-2 phenylindole.)

the right lymphatic duct. On entering the subclavian veins by way of lymphaticovenous anastomoses, the lymph is drained back into the venous system and returned to the systemic circulation. Lymphatic capillaries are thin-walled vessels that are composed of a single layer of lymphatic endothelial cells (LECs). These tiny vessels are only 30 to 80 mm in diameter, are not surrounded by pericytes or smooth muscles cells, and usually lack a basement membrane (Fig. 4-3). Lymphatic capillaries are discontinuous and contain gaps that make them highly permeable. This allows macromolecules and leukocytes to easily enter the vasculature. As lymph transits through the lymphatic capillaries, the fluid moves into precollector vessels. Unlike the capillaries, these vessels have some incomplete coverage by smooth muscle cells. The fluid then moves from precollector vessels to collecting lymphatics, which are larger vessels covered by smooth muscle cells that contain a basement membrane (see Fig. 4-3).

K23346_Neligan_04_Embryology_r5_0051-0060.indd 55

5/26/2015 7:58:57 AM

56

Part II  Anatomy, Physiology, and Lymphangiogenesis

FIG. 4-4  Collecting lymphatic vessel in a mouse hindlimb cut in a longitudinal section. Note the valve in the lymphatic vessel (black arrow).

Efferent vessels

Blood supply Hilum

Medullary sinus Marginal sinus Primary follicle

Valve

Secondary follicle with germinal center

Capsule

Trabecula

Afferent vessels

FIG. 4-5  Anatomy of a lymph node, showing afferent and efferent lymphatic vessels.

In addition, collecting lymphatics contain valves that maintain unidirectional flow (Fig. 4-4). Unlike the blood in the cardiovascular system, the lymph in the lymphatic system is not propelled forward by the heart. Instead, lymphatic fluid is moved by the relatively weak intrinsic pulsation of its smooth muscles cells in the lymphatic collectors. This force is greatly augmented by skeletal muscular contraction during periods of physical activity. As lymph travels from lymphatic capillaries back to the venous circulation, the lymph is filtered in the lymph nodes (Fig. 4-5). Lymphatic fluid enters the lymph node through afferent lymphatic vessels that drain into the subcapsular sinus and leaves the lymph node through efferent lymphatics located in the medulla. A typical adult human has 600 to 700 lymph nodes, which have various

K23346_Neligan_04_Embryology_r5_0051-0060.indd 56

5/26/2015 7:58:57 AM

Chapter 4  Embryology

57

functions. For example, lymph nodes filter out waste products found in the lymph entering from the afferent lymphatic vessel. In addition, lymph nodes provide an environment for leukocytes to interact with activated antigen-presenting cells, allowing the creation of immune responses to pathogens (see Chapter 10). This interaction is dependent on lymph node resident cells and migrating leukocytes that enter the lymph node through the high endothelial venules.

Embryology The development of the lymphatic vasculature is a stepwise process that requires the specification of LEC progenitors from the embryonic veins. These veins will give rise to the primitive lymph sacs, from which the entire lymphatic system will be derived (Fig. 4-6).

Embryonic cardinal vein

Cell differentiation Lymphatic endothelial cell progenitors Sprouting and migration

Primary lymph sac

Separation

FIG. 4-6  Stepwise embryonic development of the lymphatic vasculature.

Sprouting Primitive lymphatic vessels Merging, remodeling, maturation, and vessel differentiation Lymphatic capillary plexus

K23346_Neligan_04_Embryology_r5_0051-0060.indd 57

Lymphatic precollector vessel

Lymphatic collector vessel

5/26/2015 7:58:58 AM

58

Part II  Anatomy, Physiology, and Lymphangiogenesis

Embryology of the Vascular System The first channels of the vascular system can be seen after the third week of gestation. Vascular development is guided by receptors for growth factors. These first channels develop in three stages: • In stage 1, the undifferentiated stage, there is only a small network of capillaries. • In stage 2, the network-forming stage, plexiform structures are created that increase the system’s volume and extent. • In stage 3, the maturative stage—which occurs in the third week of gestation—the first large arteriosus, venous, and lymphatic ductus vessels stand and develop. Vessel formation—for both vasculogenesis and angiogenesis (the sprouting of blood vessels from preexisting angioblastic cords)—begins on the seventeenth day in the extraembryonic regions with the generation of blood islands in the mesoderm of the yolk sac, embryonic stalk, and chorionic villus. The vascular network expands throughout the embryonic disc, a process that gradually increases the network’s prevalence. By the twenty-fourth day, the yolk sac is connected to the embryo through two veins and three vitelline arteries (the celiac trunk, superior mesenteric artery, and inferior mesenteric artery). The red blood cells begin to circulate. The two allantoic veins and two allantoic arteries (which will become the umbilical cord) pass through the pedicle. A collection of veins, called the common cardinal veins, arise from the edge of the embryo: • Two anterior cardinal veins from the cephalic endpoint • Two posterior cardinal veins from the tail endpoint • Two common cardinal veins, left and right Initially the venous system is symmetrical, but in the second month of gestation, the right component takes over. The inferior vena cava develops from the right vitelline vein, left and right posterior cardinal veins, and superior and inferior cardinal veins. The common cardinal veins, the allantoic and vitelline veins, merge into the venous sinus, which, incorporated into the right atrium, will include the orifices of the superior and inferior vena cava. The superior vena cava develops from the common cardinal vein (right branch).

Embryology of the Lymphatic System The lymphatic system is derived in part from the mesoderm and in part from the mesenchyme, an undifferentiated tissue not yet specialized, which retains the ability to transform into another type of tissue even in adults. The mesenchymal origin of the lymphatic system is evident in the reticular cells, which constitute the frame of the immunocompetent organs, particularly the lymph nodes, which can transform into macrophages. Endothelial cells, which line the inside of the capillaries and lymph vessels, are also able to transform into macrophages. Animal model research has demonstrated that a subpopulation of blood endothelial cells, which sprout from cardinal and peripheral veins, will differentiate into LECs.2 Lymphatic vessel endothelial hyaluronan receptor 1 (LYVE1) is the earliest known cell marker indicating that LEC competence has been acquired.3

K23346_Neligan_04_Embryology_r5_0051-0060.indd 58

5/26/2015 7:58:58 AM

Chapter 4  Embryology

59

After most of the LEC progenitors bud off from the veins and migrate into the surrounding mesenchyme, lymph sacs are formed. Six primitive lymph sacs form: • Two jugular sacs, which are equal and symmetrical in cranial position, at the union of the jugular veins and anterior cardinal veins • Two iliac sacs, found at the union of the iliac and posterior cardinal veins • The retroperitoneal sac, which is located on the posterior abdominal wall, at the root of the mesentery • The cisterna chyli, positioned dorsally to the retroperitoneal bag The sacs are connected by lymphatic channels. Both main channels, born independent, are formed by primitive right and left thoracic ducts, and subsequently they join caudally with an anastomotic branch. They combine the jugular bags with the cisterna chyli. The thoracic duct of the adult grows caudally from the distal portions of the primitive thoracic ducts (right and left) and cranially from the left thoracic duct. The right lymphatic duct (large lymphatic right vein) derives from the cranial part of the right thoracic duct. The subclavian, jugular, and bronchomediastinal collector trunks derive from the jugular bags. After the primitive lymphatic vessels are formed, they will further differentiate into larger collecting vessels and smaller capillaries, which are the two predominant types of vessels of the lymphatic vasculature. In the collecting lymphatic vessels, valves form to prevent the backflow of the lymph and separate the lymphatic vessels into functional units called lymphangions. These are surrounded by smooth muscle and contract to actively transport lymph.1,4

Conclusion Clinicians must consider both the vascular and lymphatic systems as a whole and be aware of their interdependence, as seen in their clinical behavior in the adult organism. Remarkable vein malformations, such as the persistence of a marginal vein, can be associated with parallel lymphatic malformations, which must be considered in the clinical management. Lymphangiogenesis continues throughout life, influenced by growth factors and also by specific individual embryonic development of the vascular system.

R EFERENCES 1. Kiefer F, Schulte-Merker S, eds. Developmental Aspect of the Lymphatic Vascular System. Advances in Anatomy, Embryology and Cell Biology, vol 214. Heidelberg: Springer-Verlag Wien, 2014. 2. Okuda KS, Astin JW, Misa JP, et al. LYVE1 expression reveals novel lymphatic vessels and new mechanisms for lymphatic vessel development in zebrafish. Development 139:2381-2391, 2012. 3. Oliver G. Lymphatic vasculature development. Nat Rev Immunol 4:35-45, 2004. 4. Dieter R, Dieter R Jr, Dieter R III, eds. Venous and Lymphatic Diseases. New York: McGraw-Hill, 2011.

K23346_Neligan_04_Embryology_r5_0051-0060.indd 59

5/26/2015 7:58:58 AM

K23346_Neligan_04_Embryology_r5_0051-0060.indd 60

5/26/2015 7:58:58 AM

C hapter 5 Changing Concepts in Lymphatic Pathways Wei-Ren Pan

K ey P oints • A rich avalvular lymph capillary network exists in the skin and galea of the scalp, which connects adjacent lymph territories in layers.

A

• The number and diameter of the lymph collectors differ from person to person. • Some lymph collectors drain upward toward the mandible in the anterior neck above the platysma. • The lymphatic drainage patterns of the head and neck are diverse among individuals, and even within each side of the same individual.

Cha

• Knowledge of the anatomy of the human lymphatic system and advances in imaging will help clinically when surgery of any type is anticipated as well as in follow-up scenarios for patients with lymphedema.

The lymphatic system is a vast network of tiny, colorless vessels that facilitate the removal of intracellular metabolic products from the body, provide protection from disease, and are the main route of cancer metastasis, yet remains the least described in the medical literature. Our current knowledge of the lymphatic system is mostly based on investigations that were done in the nineteenth century. However, this knowledge is often discordant with clinical experience. Further, it does not explain clinical anomalies seen with melanoma and breast cancer patients using technologic developments such as lymphoscintigraphy and sentinel node biopsy for early stage cancer treatment. Importantly, it is also well known that cancers can recur at distant sites in the absence of lymph node involvement, suggesting that disseminating cancer cells can access the circulation without traversing lymph nodes. Lymphatic malformations, such as those that occur in primary lymphedemas, and damage or destruction of them cause many pathologic states, including lymphedema. However, with the application of improved knowledge and awareness, as described in this chapter, we may be able to anticipate improved outcomes for our patients. The main leverage point is that of imaging the lymphatic system, perhaps only at first in anticipated high-risk patients, preoperatively and perioperatively, and using that knowledge to vary the process or procedure to minimize lymphatic damage but at the same time not increasing the risk of cancer recurrence. Once this becomes an 61

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 61

5/27/2015 9:11:46 AM

62

Part II  Anatomy, Physiology, and Lymphangiogenesis

established procedure, this imaging could be employed for all patients whose lymphatics are about to be damaged (such as individuals who are about to undergo a surgical or radiologic intervention) or for those who already have a lymphatic malformation or defect.

History of Lymphatic Discovery The accumulation of our knowledge of the lymphatic system was a gradual one, relating mainly to improvements in techniques to help visualize the delicate and mostly transparent lymphatic vessels. Originally, cadaveric dissection and observation were the major techniques. These were used for studies of the parietal and visceral tissues of the body after Aselli identified the lymphatics in a postprandial canine in 1622.1-3 In the following years, the discovery of mercury as an imaging aid, by Nuck in 1692,4 made it possible to map the lymphatics of the body.5-7 However, it was Sappey8 who published the definitive work using mercury injection to identify the fine lymphatic networks, long before the discovery of x-rays. His artistic drawings were based on studies of many subjects and resulted in the production of an overall map of the lymphatic network of the human body. Thereafter different types of injection materials, including dyes, iodine, milk, and radioactive isotopes were substituted for the toxic mercury, and it is these which formed the basic knowledge of the lymphatic system of the body.3,9-11 However, in many ways this was still inadequate; our knowledge of the lymphatic system needed to be updated to explain some of the unexpected findings frequently seen clinically.12-14 In the past decade a series of lymphatic studies in human cadavers have been published by using a new technique with radiopaque materials as the injection medium.15-28 These results might help to clarify some unexpected findings in the clinic. Recent clinical experience in melanoma and breast cancer and the most recent cadaveric results have led to a fundamental reevaluation of the “classic” theory, since the drainage varies from the traditional predictions of Sappey and others in many cases.12,13 Therefore a more precise and eventually an individualized map of the anatomic details of the lymphatic pathways of the human body, especially in the head and neck, can form an important basis for improved clinical management of trauma, infection, lymphedema, and cancer. In the past decade we studied a total of 62 specimens harvested from 27 unembalmed cadavers (12 male and 15 female cadavers), aged between 69 and 98 years (average 86 years).16-28 The investigation commenced at the distal end of each specimen; 0.5 ml of 6% hydrogen peroxide mixed with blue drawing ink (ratio 20:1), or 6% hydrogen peroxide only, was injected into the dermis. The epidermis and dermis were incised carefully and gently under a surgical microscope. The distended lymphatic vessel was identified in the subcutaneous tissue. A 30-gauge needle was then inserted into the vessel, and a radiopaque mixture (lead oxide or barium sulphate, milk powder, and water) was injected into the vessel. If the caliber of the vessel was smaller than 0.2 mm, a glass needle (premade using thin-walled G-100 glass tubes and the Puller PP-830) was used for injection. The specimen was radiographed using a Fuji FCR IP Cassette and a Fuji Computed Radiography Processor to locate the proximal extent of the injection for further cannulation and injection. All vessels were traced, photographed, and radiographed to demonstrate the lymphatic routes from the distal side to the proximal lymph nodes.16-28

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 62

5/27/2015 9:11:46 AM

Chapter 5  Changing Concepts in Lymphatic Pathways

63

Head and Neck The drainage patterns of the head and neck were found to be different in individual specimens and even asymmetrical between the sides of the same body. The lymph-collecting vessels are dense in the scalp and lateral neck but sparse in the face and anterior and posterior neck areas. Imaging revealed a complete lymphatic map of the integument of the head and neck. The key outcome of these new observations is that the major lymphatic vessels may drain to different lymph nodes, even if they come from the same group. For example, the lymphatics of the parietal region may drain into preauricular, retroauricular, infraauricular, or even internal jugular lymph nodes. Therefore some lymph nodes could be first-tier nodes for one group of vessels but second-, third-, or even fourth-tier nodes for the other vessels. Furthermore, two layers of lymphatic vessels were found in the anterior superficial neck. Importantly, vessels in the head and neck did not always enter the closest lymph nodes but sometimes bypassed them (Figs. 5-1 and 5-2). Lymphatic vessels in the superficial tissue of the head and neck are distributed through the scalp (between the anterior hairline and occipital hairline), the face (between the anterior hairline and

Platysma Groups of lymphatic vessels: Frontal Parietal Occipital Eyelid

Nasal and oral Mental Internodal Anterior cervical (above the platysma)

FIG. 5-1  The lymphatic distribution of the superficial tissues in the head and neck (above the platysma) of one subject. Lymphatic vessels have been color coded to show each group entering their related first-tier lymph nodes.

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 63

5/27/2015 9:11:47 AM

64

Part II  Anatomy, Physiology, and Lymphangiogenesis

Groups of lymphatic vessels: Frontal Parietal Occipital Eyelid Nasal and oral Mental Internodal Anterior cervical Supraclavicular Supratrapezoid

FIG. 5-2  The lymphatic distribution of the superficial tissues in the head and neck (below the platysma). Vessels are color coded to highlight the different groups and lymph nodes.

the inferior board of the mandible), and the neck (below the inferior board of the mandible and occipital hairline) regions.

Scalp Region Our findings in the scalp showed rich avalvular lymph capillaries originating from both the dermis and the galeal layers (Fig. 5-3), where they converged to precollectors and directly drained to the collector. An alternative presentation occurred in which precollectors arose from the lymph capillaries of the dermis, crossed the subcutaneous tissue, and passed other collectors to join a precollector network in the galeal layer. Thus these were named indirect precollectors or bridge precollectors (Figs. 5-4 and 5-5). Measuring an average of 0.25 mm, the collecting lymphatic vessels are dense in the scalp region and run obliquely down to reach their first-tier lymph nodes (Figs. 5-4 and 5-6). Vessels diverged and converged along their course, like the branches of a river. Sometimes they crossed over each other and/or anastomosed with neighboring vessels. During their course, lymph collectors ran caudally in the deep aspect of the subcutaneous tissue, receiving precollectors on the way, and then drained toward their first-tier lymph nodes. The scalp lymphatic vessels include the frontal, parietal, and occipital groups, which are described next.

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 64

5/27/2015 9:11:47 AM

65

Chapter 5  Changing Concepts in Lymphatic Pathways

A

B Capillary network

Precollecting vessels

Collecting vessel 1 mm

FIG. 5-3  A, Lymph capillary network in the dermis filled with an india ink mixture. B, Lymphatic vessels in the galeal layer filled with the india ink mixture.

Initial lymphatic capillary plexus in galeal layer

Subcutaneous tissue Dermis

Lymphatic precollector vessel

Galea aponeurotica

Lymphatic collector vessels

Ampulla structures

Initial lymphatic capillary plexus in dermis

Ampulla structures

First-tier lymph node

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 65

FIG. 5-4  Basic lymphatic pathway in the scalp. The initial capillary lymphatic network originates from both the dermis and galeal layers to join a precollecting vessel that runs either directly or indirectly a short distance in both layers before forming a collecting vessel in the subcutaneous layer. Along its course, the collecting vessel divides and converges and may form different-shaped lymphatic ampullae before finally reaching the lymph node.

5/27/2015 9:11:49 AM

66

Part II  Anatomy, Physiology, and Lymphangiogenesis

A

FIG. 5-5  A, The lymph capillary network in the dermis of the scalp filled with a lead oxide mixture. B, The lymph capillary network of the skin is shown with an indirect precollecting lymph vessel that crosses the subcutaneous tissue and runs toward the galea, where it converges with the other precollectors that drain into the lymph-collecting vessel shown. C, The lymphatics are viewed from the galeal layer.

C

B

Indirect precollecting lymph vessel

Collecting lymph vessel

Skin Direct precollecting lymph vessel

Subcutaneous tissue Galeal layer (aponeurosis)

Precollecting lymph vessels

Precollecting vessel arises from the dermis piercing the subcutaneous tissue to the galeal layer

Frontal Group  Here, an average of 4 branches (range 3 to 6) of frontal collecting lymph vessels were found. These vessels course radially toward their first-tier lymph nodes in the deep aspect of the subcutaneous tissue between the superior verge of the eyebrow and the coronal suture. They drained to one or multiple lymph nodes in the preauricular, retroauricular, and deep parotid groups. Occasionally one vessel, arising from the midfrontal region, traveled anteriorly and passed over the forehead, inner canthus, and root of the nose before draining to either the nasolabial or buccinator lymph nodes. Parietal Group  Here an average of 6 branches (but with a wide range of 4 to 12) of parietal collecting lymph vessels were found in each specimen. These vessels traveled radially in the deep aspect of the subcutaneous tissue between the coronal suture and the lambdoid suture. They did not only drain to the retroauricular but also the preauricular lymph nodes, sometimes even the occipital and jugular lymph nodes. Occipital Group  Here an average of 6 (range 4 to 9) occipital collecting lymphatic vessels were found. Vessels traveled radially in the subcutaneous tissue between the lambdoid suture and the posterior hairline. They drained into the superficial, deep occipital, or jugular lymph nodes.

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 66

5/27/2015 9:11:49 AM

67

Chapter 5  Changing Concepts in Lymphatic Pathways

es ch an r b tal on r F

es ch an r b tal on r F

Preauricular lymph node

Nasolabial lymph node

Retroauricular lymph node etal Pari

tal

n Fro

Parotid lymph nodes

Preauricular lymph nodes Parotid lymph nodes

ches

Buccinator lymph node

Preauricular lymph node

Pari e

es

tal b ranc h

anch al br t e i r Pa

bran

es

ch

n bra

es

Retroauricular lymph node Retroauricular lymph nodes

Lateral internal jugular lymph nodes

Retroauricular lymph nodes

cip ita lb ran c Oc

Occipita

ches al bran Occipit

he s

Preauricular lymph nodes

Deep occipital lymph nodes

es l branch

Deep occipital lymph nodes

Superficial occipital lymph nodes

Superficial occipital lymph nodes Lateral internal jugular lymph node

FIG. 5-6  Lymphangiograms of the head and neck after lymphatic contrast injection. The frontal lymph vessels (top), parietal lymph vessels (middle), occipital lymph vessels (bottom), and related lymph nodes (purple) are highlighted.

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 67

5/27/2015 9:11:53 AM

68

Part II  Anatomy, Physiology, and Lymphangiogenesis

A

B

C

FIG. 5-7  A, Image magnified from the purple boxed area. B, The site of lymphaticovenous anastomoses located in the occipital area (black arrows). It confirms the clinical findings of Wallace et al. C, Drawing of the same area. (Green, Lymphatic vessels; blue, vein.)

Lymphaticovenous Shunt  One lymphaticovenous shunt was found in the occipital region. The efferent lymph vessels of the superficial occipital lymph nodes formed a lymphatic network. From the network two vessels emerged to communicate with a superficial occipital vein in the subcutaneous tissues (Fig. 5-7). This confirmed the clinical findings described by Wallace et al.29

Facial Region Lymphatic vessels in the face were sparse. An average of 4 branches (range 3 to 5) was identified. The average diameter of vessels was 0.3 mm when distended with injectant, but this could be partly artifactual as a result of the vessels being abnormally stretched. Vessels traveled radially from medial to lateral in the deep aspect of the subcutaneous tissue between the eyebrow and the inferior border of the mandible toward their first-tier lymph nodes. Four groups of vessels are described according to their origin (Fig. 5-8): the eyelid group, nasal group, oral group, and mental group. Eyelid Group  The initial lymphatic capillaries arose in the upper and lower eyelids (Fig. 5-9). Outer Canthus Branch  At the outer canthus of the eyelid, one to three lymph-collecting vessels were identified that then merged into a main collector that ran obliquely in the subcutaneous tissue and drained to the submandibular, or parotid lymph nodes. Inner Canthus Branch  At the inner canthus of the eyelid a collector, formed by the initial lymphatic capillaries arising in the upper and lower eyelids, ran obliquely in the subcutaneous tissue and drained to the submandibular, parotid, or buccinator lymph nodes.

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 68

5/27/2015 9:11:53 AM

69

Chapter 5  Changing Concepts in Lymphatic Pathways

Inner eyelid branch

Outer eyelid branches

Inferior eyelid branch

Outer eyelid branches

Inner eyelid branch

Inner eyelid branches Outer eyelid branches

Inferior eyelid branch

Parotid lymph nodes

Inferior eyelid branch Buccinator lymph node

Submandibular lymph node

Submandibular lymph nodes

Submandibular lymph nodes

Frontal branch

Nasal branches

Buccinator lymph nodes

Buccinator lymph node

Nasal branch

Nasal branch Oral branch

Oral branch

Submandibular lymph nodes Inner eyelid branch

Nasal branch

Nasal branch Nasolabial lymph node

Oral branch Submandibular lymph node

Oral branch Submandibular lymph node

FIG. 5-8  Lymphangiograms of the face after injection of a lymphatic contrast medium. The lymph vessels of the eyelids (top), nasal lymph vessels (middle), oral branch (bottom), and related lymph nodes (purple) are highlighted.

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 69

5/27/2015 9:11:56 AM

70

Part II  Anatomy, Physiology, and Lymphangiogenesis

Outer canthus lymphatic branch

Inferior eyelid lymphatic branch

Inner canthus lymphatic branch

Buccinator lymph node

Preauricular lymph node Parotid lymph node Submandibular lymph node

FIG. 5-9  Lymphatic drainage of the eyelids. Note that the inferior lymphatic branch of the lower eyelid can converge to either the outer or inner canthus lymphatic branches. Arrows indicate the direction of lymph flow.

Inferior Eyelid Branch  One vessel, arising from the middle-inferior aspect of the lower eyelid, ran obliquely down in the subcutaneous tissue. It merged either with the outer or inner canthus branches and drained to the submandibular or deep parotid lymph nodes. Occasionally an upper-inner canthus branch ran horizontally and laterally above the superior verge of the eyebrow and passed obliquely downward and posteriorly at the lateral edge of the eyebrow. Then it passed over the zygomatic process and descended anteriorly to converge with the main branch of the outer canthus draining to the submandibular lymph node. Nasal Group  The lymph vessel arising on the lateral side of the external nose traveled obliquely down from the median to the lateral side in the subcutaneous tissue of the cheek and drained into the nasolabial, buccinator, or submandibular lymph nodes. Oral Group  The initial lymphatic vessels arising in the perilabial tissue formed one or two collecting vessels near the corner of the mouth and drained to the buccinator, submandibular, or submental lymph nodes. Mental Group  The mental vessels ran in the deep aspect of the subcutaneous tissue and drained to the submental or submandibular lymph nodes (Fig. 5-10).

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 70

5/27/2015 9:11:57 AM

71

Chapter 5  Changing Concepts in Lymphatic Pathways

A

Submandibular lymph node

Mental branch

Submental lymph node

Superficial anterior cervical lymphatic branches

B

Anterior cervical lymphatic branches

Submental lymph node

Mental branches

Submandibular lymph node

Superficial anterior cervical lymphatic branches

C Mental branches

Platysma

Thoracic duct

Supraclavicular node

FIG. 5-10  A, Superficial lymphatic drainage of the neck. B, Radiograph shows the pathways of the anterior cervical lymphatic branches (red) situated between the dermis and platysma in the anterior neck and their related lymph nodes (purple). Green arrows indicate the direction of lymph flow. C, Lymphangiogram of the face and neck after lymphatic contrast injection, showing the mental lymph vessels (aqua blue), the anterior cervical branches (red) below the platysma, and their related lymph nodes (purple).

Anterior cervical lymphatic branches

Supraclavicular node

Thoracic duct and ampulla

Cervical Region These vessels included the anterior, lateral, and posterior groups; these are discussed next.

Anterior Cervical Group Two layers of lymphatic vessels were found in the anterior superficial neck. Above the platysma vessels traveled upward, horizontally, or obliquely. The diameter of these vessels was small, 0.1 mm to 0.2 mm. Medially, they pierced the platysma near the midline, draining into the submental lymph node between the inferior border of the mandible and the laryngeal prominence and into the supraclavicular lymph nodes between the laryngeal prominence and jugular notch. Laterally, they turned over the lateral border of the platysma and drained to the submandibular lymph nodes. The vessels below the platysma were seen running above the deep fascia. They drained to the anterior jugular lymph node and/or supraclavicular lymph node.

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 71

5/27/2015 9:11:58 AM

72

Part II  Anatomy, Physiology, and Lymphangiogenesis

FIG. 5-11  Lymphangiogram of the head and neck following lymphatic contrast injection, showing the internodal lymph vessels (blue), supratrapezoid branches (green), supraclavicular branch (yellow), and related lymph nodes (purple).

Supratrapezoid branches

Left lymphatic duct with its ampulla

Supraclavicular lymph nodes

Supraclavicular branch

Lateral Cervical Group The vessels in the lateral cervical area were numerous and complex, located in the region between the inferior ear and root of the neck and running in different directions and in different layers: the subcutaneous (superficial), intermuscular (middle), and perivascular (deep) layers. Most of these vessels were situated between lymph nodes and are called the internodal lymph-collecting vessels (Fig. 5-11).

Posterior Group The vessels of the posterior group included the supraclavicular and supratrapezoid branches. Supraclavicular Branch  The diameter of these vessels was approximately 1 mm at least when distended with the injectant. The vessels ran anteromedially in the deep aspect of the subcutaneous tissue in the root of the neck, draining to the lateral internal jugular and/or supraclavicular lymph nodes. Supratrapezoid Branch  The diameter of these vessels also averaged 1 mm. They ran anteromedially in the deep aspect of the subcutaneous tissue in the root of the neck, draining to the supraclavicular lymph nodes.

External Ear Four groups of lymph-collecting vessels were found in the external ear: the anterior, superior, middle, and inferior (lobule) branches (Figs. 5-12 and 5-13).

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 72

5/27/2015 9:11:59 AM

73

Chapter 5  Changing Concepts in Lymphatic Pathways

Superior branches

Superior branches

Middle branches

Middle branch Anterior branch

Point at which lymph vessel turns to back of ear

Anterior branch

Point at which vessel turns to back of ear

Preauricular lymph node Inferior (lobule) branches

Preauricular lymph node Infraauricular lymph node

Substernocleidomastoid lymph node

Inferior (lobule) branches

Infraauricular lymph node

FIG. 5-12  Lymphangiogram of two external auricles after injection of a lead oxide medium. The lymphatic pathways originating from different parts of the ear are highlighted with different colors.

Superior branch Middle branch Middle branch Anterior branch Preauricular lymph node Inferior branch

Substernocleidomastoid lymph node

Infraauricular lymph node

FIG. 5-13  Anterior and posterior diagrams show the lymphatic drainage of the external ear.

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 73

5/27/2015 9:12:02 AM

74

Part II  Anatomy, Physiology, and Lymphangiogenesis

Anterior Group  The initial lymphatic capillary network was distributed over most of the anterior aspect of the external ear. Medially, they converged to become one lymph-collecting vessel running under the skin of the crus of the helix and draining directly or indirectly (having merged with a vessel descending from the scalp) into the preauricular lymph node (see the green vessels in Figs. 5-12 and 5-13). Superior Group  Lymphatic vessels (here collectors are meant), arising in the superior part of the helix, traveled in the subcutaneous tissue of the back of the ear and merged together and then ran in the subcutaneous tissue of the upper lateral neck (sometimes they divided into branches) to reach the infraauricular and/or substernocleidomastoid lymph nodes (see the orange vessels in Figs. 5-12 and 5-13). Middle Group  These lymphatic vessels (here collectors are meant) arose from the scaphoid fossa near the auricular tubercle. They traveled down and passed over the cartilage at the middle of the rim and then ran obliquely in the subcutaneous tissue of the back of the ear. Continuing their course in the subcutaneous tissue of the upper lateral neck, they entered the infraauricular lymph node (see the yellow vessels in Figs. 5-12 and 5-13). Occasionally the vessel was divided before entering the infraauricular lymph node. One branch entered the node, and the other one bypassed the node and continued its course (see the large white arrow in Fig. 5-12). Inferior (Lobule) Group  Lymphatic vessels (here collectors are meant) arose in the lobule of the auricle, converged and ran obliquely down to reach the infraauricular lymph node (see the blue vessels in Figs. 5-12 and 5-13). Occasionally the vessel divided into two branches before entering the infraauricular lymph node. One entered the node while the other bypassed the node, continuing its course.

Clinical Applications of the Anatomy of the Head and Neck Rhytidectomy The postoperative incidence of prolonged edema following rhytidectomy procedures is very low but remains a frustrating complication for the surgeon and patient alike. Baker et al,30 after reviewing 1500 cases, reported that five patients had prolonged edema following rhytidectomy but did not mention any possible links between the lymphatic drainage and prolonged edema.31 Guy et al32 stated that postoperative persistent edema associated with rhytidectomy was unusual and presumably was related to lymphatic stasis, but they did not provide further discussion, and details of the lymphatic anatomy in this region were inadequate. Because the lymphatic drainage patterns of the superficial tissue in the head, face, and neck are different in each individual, the incision of the rhytidectomy might cross lymph-collecting vessels that gather in the preauricular area in some cases (Fig. 5-14) and cross those located in the subcutaneous tissue of the dissection area. Overdissection in this region might disrupt the major lymphatic vessels and result in the frustrating complication of edema. Lymphedema of the eyelid is an uncommon condition that presents in many cases as a chronic form related to acne, rosacea, irradiation, and ocular surgery.33 It has been mentioned that the degree of swelling is usually related to surgical factors such as excessive cauterization, extensive tissue manipulation or excision, and dissection in the lateral canthal area resulting in lymphatic interruption.34 Fig. 5-9 demonstrates that the avalvular initial lymphatic capillaries arose in the up-

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 74

5/27/2015 9:12:02 AM

Chapter 5  Changing Concepts in Lymphatic Pathways

75

FIG. 5-14  The relationship of the facial lymphatics and the part of the preauricular incision in rhytidectomy.

per and lower eyelids where the lymph could flow to the outer-inner canthus or inferior branches. If one of those branches was damaged or blocked, the others could still perform the function. It is likely that severe edema would occur if all branches were damaged by a severe injury of the periocular soft tissue or by multiple surgical incisions on the eyelid.35

Sentinel Lymph Node Mapping and Biopsy Lymphoscintigraphy is an important diagnostic technique used to identify the sentinel lymph node.12,13 Sentinel lymph node biopsy has become an important procedure in the treatment of patients with melanomas in the head and neck.13 However, lymphoscintigraphy results13 have often been discordant with clinical predictions based on our “classic” knowledge of the lymphatic system. It was found that the false-negative result could be as high as 34% in patients with skin cancer of the head and neck area.13 This percentage suggests that the surgeon will fail to remove nodes potentially containing metastatic disease in one of three patients. The findings indicated in this section will help clinicians to reevaluate the often unexpected findings in lymphoscintigraphic results.16,19,22,23,25-27 Although the lymphatic anatomy of the head and neck, with the first-tier lymph nodes, have been described and demonstrated by photographs and radiographs, several important points still need to be emphasized: • A rich avalvular lymph capillary network exists in the skin and galea of the scalp, which connects adjacent lymph territories in layers. • Indirect precollector lymph vessels link up to the lymph capillaries of the skin and galea in the scalp. • The number and diameter of the lymph collectors differ from person to person.

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 75

5/27/2015 9:12:03 AM

76

Part II  Anatomy, Physiology, and Lymphangiogenesis

• Some lymph collectors drain upward toward the mandible in the anterior neck above the platysma. • The lymphatic drainage patterns of the head and neck are diverse among individuals, and even within each side of the same individual. • The lymphatic vessels of the head and neck do not always enter the first-tier (or the nearest) lymph nodes but sometimes bypass them. Some of the lymphatic characteristics listed above may be unique to the head and neck section, and some may be common features in other parts of the body, as will be described in the following sections.

Breast and Anterior Torso Two layers and three groups of lymphatic collectors of the female breasts and anterior torso were identified according to their origins. The superficial layer includes the subareola and superficial tissue groups; the deep layer contains the internal thoracic vascular group. Lymph collectors were found evenly spaced in the superficial layer of the anterior upper torso, draining radially into the axillary lymph nodes. The patterns of the drainage territories of the firsttier lymph nodes varied among specimens and even between the sides of the same subject (Fig. 5-15), as was similarly described for the head and neck areas. As vessels reached the breast, some passed over and some through the breast parenchyma. In one case, one lymph node in the axilla drained almost the entire breast. In the majority, however, more than one node was represented (Fig. 5-16).18 Sometimes one large axillary lymph node drained the anterior chest and breast as well as most of the medial surface of the upper limb (Fig. 5-17).17

LN1 LN1 LN2

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 76

LN2

FIG. 5-15  The asymmetrical pattern of drainage is evident. White arrows indicate two lymphatic collectors arising from each side of the nipple-areola complex. The yellow arrow indicates the lymphatic plexus in the periareolar region. The green arrow indicates the vessel from the superficial to the internal mammary lymphatic system. Black arrows indicate the nipple-areola complex. The green lymphatic vessels drain to both lymph nodes 1 and 2; yellow vessels draining to lymph node 1 on the left and 2 on the right; the red vessels are the internal mammary lymphatic system. (LN, Lymph node.)

5/27/2015 9:12:04 AM

Chapter 5  Changing Concepts in Lymphatic Pathways

A

B

C

D

77

FIG. 5-16  Tracing distally of lymphatics of both hemi-upper torsos (A and C are male; B and D are female) from each firsttier lymph node color coded; pectoral node (green, orange, black, and yellow ), subclavicular node (light blue), and internal mammary node (red).

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 77

5/27/2015 9:12:05 AM

78

Part II  Anatomy, Physiology, and Lymphangiogenesis

Subclavian vein

Axillary vein

Axillary lymph node draining breast and medial upper limb

Thoracodorsal vein

FIG. 5-17  An axillary node can drain the entire breast and a large area of the upper limb.

Subareola Group On average, two precollecting lymph vessels arose from the subareola plexus of each breast (see the yellow vessels in Fig. 5-15). They converged with the superficial lymphatic vessels of the anterior upper torso and coursed radially toward the axillary lymph node (or nodes).

Superficial Tissue Group The superficial lymphatic vessels arose in the subcutaneous tissues around the costal margin and the lateral border of the sternum. They coursed radially toward their first-tier lymph nodes (axillary lymph nodes). Most vessels converged to form larger lymph collectors, and some diverged before entering their lymph nodes. The size and number of vessels varied between sides, demonstrating the asymmetrical drainage patterns between the two breasts. In both the left and right axillae, all the vessels drained into one or two different sized axillary lymph nodes (see the green vessels in Fig. 5-15).

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 78

5/27/2015 9:12:06 AM

Chapter 5  Changing Concepts in Lymphatic Pathways

79

Internal Thoracic Vascular Group One or two precollecting lymph vessels arose in the inner costal margin behind rectus abdominis on each side of the chest. They converged with the superficial lymphatic vessels of the anterior upper torso and course upward to reach their first-tier lymph node and then pass through several lymph nodes in a row to emerge with the lymphatic vessel from the supraclavicular area (see the red vessels in Fig. 5-15).

Clinical Implications: Lymph Collectors of the Breast and Anterior Torso Sentinel Lymph Node Mapping and Biopsy The treatment of breast cancer has been one of the most rapidly changing fields in surgery. For example, developments in the sentinel lymph node biopsy concept are now widely applied for the treatment of patients with breast cancer, especially in its early stages. The information provided in this section is pertinent to breast cancer research and clinical applications. The findings highlight several key points regarding the use of sentinel lymph node mapping and biopsy: our finding of varying patterns of lymphatic drainage between contralateral sides indicates that an individualized approach to each breast is essential. Intraparenchymal lymphatics course superficially and thus are amenable to surgical approaches. Lymphatics do coalesce at the nipple; however, many lymphatic vessels bypass the nipple, and thus a periareolar injection (rather than peritumoral injection) of nuclear tracer or dye may not map the appropriate lymphatics. The anatomy of lymph collectors in the breasts, anterior upper torso, and the internal thoracic vascular bundles has been revealed. The drainage patterns of lymphatics into axillary lymph nodes has been demonstrated, with a predominance of superficial lymphatics, but with some parenchymal lymphatics identified. Once again, the patterns of lymphatic vessels found from origins to the first-tier lymph node in the region were asymmetrical between each side of the breasts even within an individual. Furthermore, sometimes one large axillary lymph node drained the anterior chest and breast as well as most of the medial surface of the upper limb.

Upper Limb Breast cancer–related lymphedema of the upper extremity is to date an unsolved complication that affects large numbers of patients after breast cancer surgery. The reported incidence of upper limb lymphedema in breast cancer survivors has ranged from 2% to 5% after sentinel lymph node biopsy,36,37 and 9% to 41% after axillary lymph node dissection.38 Detailed identification of lymphatic anatomy of the upper limb will help the clinician manage the lymphedema. Multiple lymph collectors were identified in the subcutaneous tissue of the upper limb, originating beneath the dermis in the midaxial lines of the fingers, wrist crease, and lateral side of the upper arm (Fig. 5-18).

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 79

5/27/2015 9:12:06 AM

80

Part II  Anatomy, Physiology, and Lymphangiogenesis

Digits Each digit contained two collecting lymphatic vessels, one lateral and the other medial. The diameters of the vessels varied from 0.2 to 0.5 mm (average 0.3 mm) and were larger on the proximal side and smaller on the distal end. They originated beneath the dermis alongside the distal phalanges and generally traveled tortuously along the midaxial lines in the subcutaneous tissue. Generally, the vessels of neighboring digits converged in the web spaces of the hand, the exception being those on the lateral border of the thumb and the medial border of the little finger. These then traveled radially to merge with lymphatic vessels in the dorsum of the hand (vessels colored blue and green in the fingers in Fig. 5-18). A transverse section of the finger showed that the digital lymphatic vessels traveled in the mid axial lines where they paralleled arteries, veins, and nerves (Fig. 5-19).

F

Axillary lymph nodes

Cubital lymph node

C

A

E

D

B

Palm

Thumb Little finger

FIG. 5-18  Superficial lymphatic distribution of the right upper limb. Left, Anteroposterior view; center, lateral view; right, lymphatic distribution of the integument. Each group of lymphatics was color coded. Vessels colored in green were divided by the incision. (A, Styloid process of the ulna; B, styloid process of the radius; C, medial epicondyle of the humerus; D, olecranon; E, lateral epicondyle of humerus; F, acromion of the scapula.)

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 80

5/27/2015 9:12:07 AM

81

Chapter 5  Changing Concepts in Lymphatic Pathways

A

B B LV

A

LV

T N

A

T N

TD

V

2 mm

FIG. 5-19  A, Transverse section of digital lymphatic vessels (filled with lead oxide mixture) of the right index finger. B, Transverse section of the proximal joint of the right middle finger. (A, Digital arteries [inserted metal wires]; B, head of proximal phalanx of the middle finger; LV, digital lymphatic vessels [filled with a barium sulfate mixture]; N, digital nerves; T, flexor digitorum superficialis tendon; TD, flexor digitorum profundus tendon; V, digital veins.)

FIG. 5-20  The relationship of the superficial lymphatic vessels (filled with a barium sulfate and dye mixture) and veins on the dorsum of the right hand.

Hand An average of 16 lymph collectors as the continuation of lymph vessels arising from digits (range 14 to 18) were distributed in the subcutaneous tissue of the dorsum of the hand. The diameters of the vessels varied from 0.2 to 0.6 mm (average 0.4 mm). Vessels diverged and converged to each other, and crossed over or below the superficial veins when they met (Fig. 5-20). They formed two groups: dorsoradial and dorsoulnar.

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 81

5/27/2015 9:12:08 AM

82

Part II  Anatomy, Physiology, and Lymphangiogenesis

Dorsoradial Group  Lymph collectors arising from the three digits were distributed on the radial side of the dorsum (see the green vessels in Fig. 5-18). Dorsoulnar Group  Lymph collectors arising from the ulnar two digits distributed on the ulnar side of their dorsum (see the blue vessels in Fig. 5-18).

Forearm An average of 24 lymph collectors (range 17 to 30), distributed in the subcutaneous tissue of the forearm region with the diameters of the vessels varied from 0.3 to 0.6 mm (average 0.5 mm) were found. They were dense and paralleled the cephalic and basilic system of veins mostly on the surface of the forearm, except the dorsum of the elbow. Three groups of lymph collectors were identified: radial, ulnar, and volar. Radial Group  Arising from the dorsoradial group of the hand, these lymph collectors traveled in dorsoventral direction and crossed the radial forearm margin from the dorsoradial side of the wrist to the lateral part of the cubital fossa (see the green vessels in Fig. 5-18). Ulnar Group  Arising from the dorsoulnar group of the hand, these lymph collectors traveled dorsoventrally and crossed the ulnar forearm margin from the dorsoulnar side of the wrist to the medial part of the cubital fossa and the elbow (see the blue vessels in Fig. 5-18). Volar Group  Arising near the wrist crease, these lymphatic collectors traveled in the subcutaneous tissue of the volar of the forearm toward the cubital fossa (see the yellow vessels in Fig. 5-18). Clustering in the anteromedial aspect of the elbow, these three groups of collectors paralleled basilic veins and crossed over and below the median and basilic veins when meeting them. In

LN

V

N

E

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 82

FIG. 5-21  The relationship of the superficial lymphatic vessels (filled with a barium sulfate mixture), veins, and nerve on the left cubital fossa. (E, Medial epicondyle of humerus; LN, lymph node; N, medial cutaneous nerve; V, basilic vein.)

5/27/2015 9:12:09 AM

Chapter 5  Changing Concepts in Lymphatic Pathways

83

some cases, a cubital lymph node presented next to the basilic vein in the cubital fossa connected by a single afferent and efferent lymph collector, while the other vessels bypassed it (Fig. 5-21).

Upper Arm An average of 19 lymph collectors (range 17 to 21), were present in the upper arm. The diameters of these collectors varied from 0.3 to 1.2 mm (average 0.6 mm). Above the elbow, the collectors were dense on the medial side and sparse on the lateral side. They converged centrally to form larger vessels before entering the lymph nodes in the axilla. There was a definite tendency for postaxial clustering and increased vessel diameter in this region. Medial Group  Clustering above the elbow level in the medial side of the upper arm, three groups of the lymph collectors from the forearm converged to form larger diameter collectors before entering the lymph nodes in the axilla (see the vessels colored green, yellow, and blue in Fig. 5-18). Two or three vessels accompanied the basilic vein and medial cutaneous nerve (Fig. 5-22). Anterolateral Group  Lymph collectors originated from the lateral side of the upper arm and traveled obliquely and horizontally via the volar aspect of the upper arm to merge with the medial group of vessels before entering the axillary lymph node (see the brown vessels in Fig. 5-18). Posterolateral Group  Originated from the lateral side of the upper arm, vessels traveled obliquely and horizontally along the dorsal aspect to merge with the medial group of vessels before entering the axillary lymph node (see the brown vessels in Fig. 5-18).

LN3 LN2

N

V

FIG. 5-22  The relationship of the superficial lymphatic vessels (filled with a barium sulfate mixture), veins, and nerve on the medial aspect of the left upper arm. (E, Medial epicondyle of the humerus; LN1, cubital lymph node; LN2 and LN3, axillary lymph nodes; N, medial cutaneous nerve; V, basilic vein.)

LN1

E

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 83

5/27/2015 9:12:09 AM

84

Part II  Anatomy, Physiology, and Lymphangiogenesis

Clinical Implications of the Anatomy of the Upper Limb Secondary lymphedema of the upper limb is one possible sequela of breast cancer surgery. This unsolved iatrogenic complication is characterized by a progressive pathologic condition in which there is an interstitial accumulation of the protein-rich lymphatic fluid, recurrent cellulitis, hypertrophy of adipose tissue, and fibrosis, resulting in increased lymph volume and chronic edema in the affected region. Creation of a lymphaticovenous anastomosis is one of the contemporary options for surgical treatment of secondary lymphedema. Detailed lymphatic anatomy of the upper limb may help to locate the lymphatic collecting vessels for the proposed surgical procedure. In summary, the characteristic lymphatic distribution of the upper limb has been described as follows: • Lymph collectors were dense in the dorsum of the hand, forearm, and medial aspect of the upper arm. • Lymph collectors could be found surrounding the basilic and cephalic veins with their accessories. • The largest vessels were located in the medial aspect of the upper arm. Awareness of this information should emphasize to surgeons the importance of searching for the location of the best lymphatic collectors for potential lymphaticovenous anastomosis sites in the upper limb.

Lower Limb Multiple lymph collectors were identified in the subcutaneous tissue of the lower limb (Fig. 5-23). They originated beneath the dermis of each side of the toes, the foot, and the lateral side of the thigh. The diameters of the vessels varied from 0.2 to 2.2 mm. The vessels traveled in a tortuous fashion through the subcutaneous tissue toward the lymph nodes in the popliteal fossa, those adjacent to the superficial femoral vessels, and those in the inguinal region. During their course some vessels branched, diverged, or converged; sometimes they anastomosed with or crossed over neighboring vessels. As they approached the lymph node groups, most vessels converged to form larger collectors. Some of these larger collectors then split into small branches just before entering the lymph nodes. The drainage patterns were different in the individual specimens and even asymmetrical between each side of the same body. They traveled in a meandering fashion within the subcutaneous tissue and passed over and/or under the veins when they met them.

Toes Each toe contained two lymph collectors, one on each side, traveling in the subcutaneous tissue along the midaxial lines. The mean vessel diameter was 0.5 mm (range 0.2 to 0.8 mm). The vessels of neighboring toes converged in the web spaces of the foot, except for the vessels on the medial and lateral borders of the first and fifth toes. They then traveled radially to merge with lymphatic vessels in the dorsum of the foot.

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 84

5/27/2015 9:12:10 AM

85

Chapter 5  Changing Concepts in Lymphatic Pathways

DILN E

SILN

G

F

FLN

SPLN

DPLN

D C

Posterior

Anterior

A

B

Sole

FIG. 5-23  Superficial lymphatic distribution of the left lower limb. Left, Anteroposterior view; center; lateral view; right, lymphatic distribution of the integument; far right, the parafemoral lymphatic vessels. Each group of lymphatics was colored differently. Black arrows indicate the cut points of vessels. (A, Lateral malleolus; B, medial malleolus; C, lateral epicondyle of femur; D, medial epicondyle of femur; E, anterior superior iliac spine; F, pubic tubercle; G, ischial tuberosity. DILN, Deep inguinal lymph nodes; DPLN, deep popliteal lymph nodes; FLN, femoral lymph nodes; SILN, superficial inguinal lymph nodes; SPLN, superficial popliteal lymph nodes.)

Foot An average of 14 lymph collectors (range 9 to 19) were found in the subcutaneous tissue of the dorsum of the foot. The mean vessel diameter was 0.6 mm (average 0.2 to 1.2 mm). They branched, diverged, converged, and anastomosed with or crossed over neighboring vessels, forming a large lymphatic network in three groups. Anterior Group  These arose from the toes, forming the front part of the lymphatic network on dorsum of the foot (see the light blue vessels in Fig. 5-23). Medial Group  These arose from the medial border of the foot, forming the medial part of the lymphatic network on the dorsum of the foot (see the yellow vessels in Fig. 5-23).

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 85

5/27/2015 9:12:13 AM

86

Part II  Anatomy, Physiology, and Lymphangiogenesis

G S V

FIG. 5-24  Lymphatic vessels (filled with a barium sulfate mixture) of the medial group in the middle of the left leg traveling with the great saphenous vein (GSV) and its branch. Green arrows indicate the direction of lymphatic flow.

Lateral Group  These arose from the lateral border of the foot, forming the lateral part of the network on the dorsum of the foot (see the green vessels in Fig. 5-23).

Ankle An average of 12 lymph-collecting vessels (range 9 to 17) were found in the subcutaneous tissue around the ankle. The mean vessel diameter was 1.0 mm (range 0.2 to 2.0 mm). Anterior Group  Most of these vessels (mean 10; range 8 to 13) were distributed in the anterior aspect of the ankle between the lateral and medial malleoli. They were continuous with vessels from the dorsum of the foot. Posterior Group  Only a few vessels (mean 2; range 1 to 4) were distributed in the posterior aspect of the ankle, arising from the sides of the Achilles tendon (tendocalcaneus) bilaterally in the subcutaneous tissue just above the heel.

Leg An average of 13 lymph-collecting vessels (range 12 to 16) were distributed in the subcutaneous tissue of the leg. The mean vessel diameter was 1.0 mm (range 0.2 to 1.8 mm).

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 86

5/27/2015 9:12:13 AM

Chapter 5  Changing Concepts in Lymphatic Pathways

87

S S V

FIG. 5-25  Lymphatic vessels after injection of a lead oxide mixture traveling with the small saphenous vein (SSV) in the calf. Green arrows indicate the direction of lymphatic flow.

Anteromedial Group  They were denser with a straighter course, following the adjacent great saphenous vein (GSV) and its branches (see the yellow vessels in Fig. 5-23) (Fig. 5-24). Anterolateral Group  Vessels were relatively sparse, following a curving course and tending toward the anteromedial aspect of the proximal third of the leg (see the green vessels in Fig. 5-23). Posterior Group  There were only one or two large lymphatic vessels (mean diameter 1 mm; range 0.7 to 1.4 mm) in the posterior aspect of the leg accompanying the small saphenous vein (SSV) (see the orange vessels in Fig. 5-23) (Fig. 5-25).

Knee An average of 15 lymph collectors (range 11 to 17) were distributed in the subcutaneous tissue around the knee region. The mean vessel diameter was 0.8 mm (range 0.3 to 1.6 mm). Anterior Group  The anterior lymphatics were relatively sparse and traveled obliquely toward the anteromedial aspect of the thigh (see the green vessels in Fig. 5-23). Medial Group  The medial vessels were denser and were formed by the anteromedial and anterolateral groups from the leg region, following the adjacent GSV and its branches (see the yellow and green vessels in Fig. 5-23).

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 87

5/27/2015 9:12:14 AM

88

Part II  Anatomy, Physiology, and Lymphangiogenesis

DPLN

SPLN

SPLN

S S V

FIG. 5-26  Left, Posteroanterior view of an inverted radiograph showing the lymphatic distribution in the knee region. Posterior lymph vessels are represented in orange, internodal lymph vessels are yellow, and superficial femoral lymph vessels are blue. (DPLN, Deep popliteal lymph nodes.) Right, A magnified image from the circled area in the left radiograph. Lymphatic vessels after lead oxide mixture injection around the small saphenous vein (SSV) in the popliteal fossa entering the superficial popliteal lymph nodes (SPLN). Green arrows indicate the direction of lymphatic flow.

Posterior Group  There were only one or two larger lymphatic vessels in the popliteal fossa following the SSV entering the superficial popliteal and/or then the deep lymph node (or nodes) (see the orange vessels in Fig. 5-23) (Fig. 5-26).

Thigh An average of 29 lymph collectors (range 27 to 31) were found in the thigh. The mean vessel diameter was 0.8 mm (range 0.3 to 1.7 mm). Anterior Group  Vessels originated from the anterolateral aspect of the thigh, running obliquely in the subcutaneous tissue before entering the lateral group of the superficial inguinal lymph nodes (see the pink vessels in Fig. 5-23). Medial Group  The medial group consisted of the continuous vessels of the anteromedial and anterolateral groups from the leg, running in the subcutaneous tissue of the medial aspect of the

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 88

5/27/2015 9:12:16 AM

Chapter 5  Changing Concepts in Lymphatic Pathways

89

SILN

G S V

G S V

FIG. 5-27  Lymphatic vessels after injection of a lead oxide mixture traveling with the great saphenous vein (GSV) in the calf. (SILN, Superficial inguinal lymph nodes.)

thigh. Vessels lay adjacent to the GSV and its branches and entered the center group of the superficial inguinal lymph nodes (see the yellow and green vessels in Fig. 5-23) (Fig. 5-27). Posterior Group  The posterior group originated from the posterolateral side of the thigh. Vessels ran obliquely in the subcutaneous tissue of the posterior aspect of the thigh toward the medial group of the superficial inguinal lymph nodes (see the blue vessels in Fig. 5-23).

Alternative Pathways From the Popliteal to Inguinal Lymph Nodes From the popliteal fossa to the inguinal lymph nodes, vessels traveled along three lymphatic pathways in the thigh region. Multiple pathways presented in the individual specimen (Figs. 5-28 and 5-29) include the superficial pathway, the parafemoral pathway, and the deep pathway. Other pathways were described by Viamonte and Rüttimann39: • The deep lymphatic vessels accompanying the obturator artery drained into the internal iliac lymph node. • Vessels accompanying the sciatic nerve drained into the inferior gluteal lymph node.

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 89

5/27/2015 9:12:16 AM

90

Part II  Anatomy, Physiology, and Lymphangiogenesis

DPLN V

SPLN

N MLV V

FIG. 5-28  Distribution of lymphatics in the left popliteal fossa. A vessel on the medial side (MLV) enters the superficial popliteal lymph node (SPLN); its efferent vessel continues in the subcutaneous tissue and then converges with the medial group of the thigh. A lateral lymph vessel (LLV) enters the deep popliteal lymph node (DPLN). (N, Tibial nerve in the popliteal fossa; V, branches of the small saphenous vein.)

LLV 5 mm

1 Superficial lymphatic pathway

Sciatic nerve

2 Parafemoral lymphatic pathway

3 Deep lymphatic pathway

External iliac lymph nodes (EILN) Superficial inguinal lymph nodes (SILN) Deep inguinal lymph nodes (DILN) Great saphenous vein Deep femoral vein Femoral vein Femoral lymph node (FLN) Deep popliteal lymph node (DPLN) To lumbar trunk DILN

EILN

Superficial popliteal lymph node (SPLN)

SILN

Deep popliteal vein and tibial nerve

1 2

Small saphenous vein

3

FLN DPLN SPLN From lateral heel

FIG. 5-29  Alternative pathways from the popliteal to inguinal lymph nodes.

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 90

5/27/2015 9:12:18 AM

Chapter 5  Changing Concepts in Lymphatic Pathways

91

FIG. 5-30  Afferent lymph vessel from the superficial popliteal lymph node running in the subcutaneous tissue of the anteromedial side of the thigh and entering the superficial inguinal lymph node.

Popliteal fossa Patella

Superficial Pathway  The efferent lymph vessel, from the superficial popliteal lymph node, ascended in the subcutaneous tissue, turning toward the medial side at the distal part of the thigh and then the anteromedial side at the top of the thigh to join the medial group vessels of the thigh before entering the superficial inguinal lymph node (Fig. 5-30). Parafemoral Pathway  The efferent lymph vessel, from the deep popliteal lymph node, were coursing in variable relationship to the femoral vascular bundle (anteriorly, posteriorly, laterally, and medially between the femoral artery and vein) and sometimes winding around the bundle before entering the lymph nodes in the inguinal area (see the brown vessels in Figs. 5-23, 5-29, and 5-31, A). The vessels were single, dividing into multiple vessels before entering the inguinal lymph nodes. Deep Pathway  The efferent lymph vessel, from the deep popliteal lymph node, was running deep to the anterior side of the sciatic nerve near the superior margin of the popliteal fossa, and then between the sciatic nerve and the profunda femoral vessels and the common femoral vessels draining into an anterior external iliac lymph node (Fig. 5-31).

The Depth of Lymph Vessels in the Subcutaneous Tissue The depth of lymph collectors depends on the thickness of the surrounding subcutaneous tissue. On the dorsum of the foot, the vessels ran within a very thin layer of tissue, whereas in the groin, a great number of collectors were packed within the entire layer of tissue. Kubik and Manestar40 reported that the lymphatic vessels of the thigh formed three layers: the first layer lay immediately

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 91

5/27/2015 9:12:19 AM

92

Part II  Anatomy, Physiology, and Lymphangiogenesis

A

External iliac lymph nodes

B

Sciatic nerve

Lymphatics running between sciatic nerve and profunda vessels

Deep popliteal lymph nodes

Lymphatics running between sciatic nerve and profunda vessels Lymphatic running with femoral vascular bundle

Superficial popliteal lymph nodes

Deep popliteal lymph node Superficial popliteal lymph node

FIG. 5-31  A, Radiograph and B, photograph after lead oxide mixture injection showing the lymphatics in the lower limb. In the radiographic lateral view (left) and posteroanterior view (right) and photograph, note the two deep collecting lymph vessels in the thigh region.

below the surface of the subcutaneous fat, the second layer lay between the first and third layers, and the third layer lay on the deep fascia. However, Pan et al28 found that the lymph collectors were distributed in close association with the great and small saphenous vein, traveling tortuously in different depths of the subcutaneous tissue.

Clinical Implications: The Lower Limb Secondary lymphedema of the lower limb is a common occurrence after trauma, infection, and surgery, especially after a radical clearance of the inguinal lymph nodes. The reported incidence of lower limb lymphedema after groin dissection has ranged from 40% to 67%.41 Surgical treatment for secondary lymphedema requires accurate knowledge of the anatomy of lymphatic routes in the lower limb to assist in preoperative preparation and intraoperative management, ultimately affecting the postoperative outcome. As emphasized throughout this chapter, the entire lymphatic drainage pattern of the superficial tissues, including the femoral lymph vessels, has shown that quantitative (number of vessels) and qualitative (size of vessels) variations exist among individuals. These data provide an important baseline for clinical applications. Most lymphatic vessels have been found to drain into their first-

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 92

5/27/2015 9:12:20 AM

Chapter 5  Changing Concepts in Lymphatic Pathways

93

tier lymph nodes in the groin (superficial inguinal lymph nodes). In contrast, there were one or two lymph collectors arising from the lateral side of the Achilles tendon in the subcutaneous tissue above the heel which traveled with the small saphenous vein. These vessels drained through popliteal and femoral lymph nodes before finally entering the deep inguinal, external iliac or inferior gluteal lymph node.24,39 Lymphaticovenous anastomoses have been performed to treat secondary lymphedema of the lower extremities.42-46 It has been mentioned that as many anastomoses as possible must be created to obtain the best result in this procedure.47 Figs. 5-23 through 5-31 provide a roadmap for surgeons when searching for potential lymphaticovenous anastomotic sites in the lower extremity where lymph vessels are situated close to veins, according to the following major scenarios: • The dorsum of the foot: rich lymphatic vessels were present in the same plane as the dorsal metatarsal veins (see Fig. 5-23). • The medial aspect of the lower extremity: abundant lymph vessels traveled with or crossed the great saphenous vein and its branches in the anterior tibial region adjacent to the medial malleolus, the medial leg, knee, and thigh (see Figs. 5-23, 5-24, and 5-27). • Posterior aspect of the leg: one or two large lymph vessels always accompanied the small saphenous vein (see Figs. 5-25 and 5-26). Lymphatic grafting procedures for treating secondary lymphedema have been reported.48 These procedures involve the harvest of single or multiple lymphatic vessels from the ventromedial bundle (medial group of the thigh) in the contralateral thigh (healthy limb). The distal end of these vessels in the donor side was anastomosed to vessels in the affected limb through a subcutaneous tunnel above the pubic symphysis. Alternatively, the vessels were harvested as free grafts for transfer to the upper limb.49 The results from this study have provided details of the lymphatic distribution in the thigh. If one or more vessels were harvested from the ventromedial bundle of the thigh, the remaining lymphatic vessels could probably still perform the required drainage function. Furthermore, vessels were constant and easily located, traveling on both sides of the small saphenous vein in the same depth of tissue in the posterior aspect of the leg. This could act as an additional donor site for harvesting lymphatic graft tissue for surgical treatment. The lower extremity (especially the medial thigh) is a common donor site for harvesting a flap. The details of the neurovascular supplies of these flaps have been well documented, but their lymphatic supply was not well understood.50-53 Fig. 5-23 identifies the afferent and efferent lymphatic pathways and the direction and quantitative distribution of lymph vessels within in the lower extremity to assist surgeons in designing soft tissue flaps (lymphatic bridging flaps) for surgical treatment of lymphedema patients.54-58 For example, a flap harvested from the medial aspect of the thigh would contain rich vertical lymphatic vessels, whereas a flap from lateral side of the thigh would contain fewer (obliquely oriented) vessels.

Sentinel Lymph Node Mapping and Biopsy Lymphoscintigraphy has been used to determine the sentinel lymph nodes for sentinel lymph node biopsy for the treatment of cancer patients.12-13 Reynolds et al59 created three-dimensional, colorcoded “heat maps” to show the lymphatic drainage patterns in patients with cutaneous melanoma. After analyzing lymphoscintigraphy data collected from more than 5000 patients, they found one patient with a melanoma on the right heel and lymphoscintigraphy results that showed lymphatic drainage to the ipsilateral popliteal lymph node fields, ipsilateral groin, and contralateral groin.

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 93

5/27/2015 9:12:20 AM

94

Part II  Anatomy, Physiology, and Lymphangiogenesis

This is a very surprising result, of course, but does indicate the variability in drainage pathways we are faced with when it comes to the lymphatic system. The results from this section have shown that the lymphatic drainage route from the lateral heel to the popliteal fossa passing through the subcutaneous tissue in the calf is constant. One or two collecting lymph vessels accompanying the small saphenous vein enter the popliteal lymph nodes. However, from the popliteal fossa to the groin area, there are three lymphatic pathways: through the superficial tissue of the medial side of the thigh, running with the superficial femoral blood vessels, or running between the sciatic nerve and the profunda femoral vessels. In addition to these three major routes, other pathways were described by Viamonte and Rüttimann,39 these being ones in which the deep lymphatic collectors accompanying the obturator artery drain into the internal iliac lymph node and one in which the lymph collectors accompanying the sciatic nerve drain into the inferior gluteal lymph node. These findings help explain the case reported by Reynolds et al59 (that is, drainage to the ipsilateral node fields and groin) and support their comments that lymphatic pathways from the popliteal fossa (efferent vessels of the lymph nodes) pass through different routes to reach different lymph node groups in the inguinal and pelvic areas.

Conclusion Actual and accurate lymphatic distribution and drainage patterns of the head and neck, anterior torso and breast, and extremities have been described and illustrated, as have the characteristics of the lymphatic anatomy in these regions. These empirical anatomic data provide the supplementary information to enrich our knowledge of the human lymphatic system, which may help clinically, especially when surgery of any type is anticipated, but also in follow-up scenarios when issues of lymphedema are being treated by therapists.

R EFERENCES 1. Aselli G. De Lactibus Sine Lacteis Venis. Milan, Italy, 1627. 2. Pecquet J. Experimenta Nova Anatomica. Paris, 1651. 3. Haagensen CD, Feind CR, Herter FP, et al, eds. The Lymphatics in Cancer. Philadelphia: WB Saunders, 1972. 4. Nuck A. Adenographia Curiosa et Uteri Foeminei Anastome Nova. Lugduni Betavorum, P vander Aa, 1692. 5. Cruikshank W. The Anatomy of the Absorbing Vessels of the Human Body. London: G Nicol, 1786. 6. Mascagni P. Vasorum Lymphaticorum Corporis Humani Historia et Ichonographia. Siena, Italy: P Carli, 1787. 7. Bonamy C, Broca P, Beau ME. Atlas d’Anatomie Descriptive du Corps Humain. Paris: Victor Masson, 1850. 8. Sappey PC. Anatomie, Physiologie, Pathologie des Vaisseaux Lymphatiques. Paris: Adrien Delahaye, 1874. 9. Gerota D. Zur Technik der Lymphgefassinjection. Eine neue Injections-masse für Lymphgefasse. Polychrom Injection. Anat Anzeiger 12:216-224, 1896.

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 94

5/27/2015 9:12:20 AM

Chapter 5  Changing Concepts in Lymphatic Pathways

95

10. Bartels P. Das lymphgefassystem. In Bardeleben K, ed. Handbuch der Anatomie des Menschen (4th part). Jena: Gustav Fischer Verlag, 1909. 11. Rouvière H. Anatomy of the Human Lymphatic System. Michigan: Edwards Brothers, 1938. 12. Uren RF, Thompson JF, Howman-Giles RB. Lymphatic Drainage of the Skin and Breast. Sydney, Australia: Harwood Academic Publishers, 1999. 13. Thompson JF, Morton DL, Kroon BBR. Textbook of Melanoma. London: Martin Dunitz, 2004. 14. Liu NF, Lu Q, Jiang ZH, et al. Anatomic and functional evaluation of the lymphatics and lymph nodes in diagnosis of lymphatic circulation disorders with contrast magnetic resonance lymphangiography. J Vasc Surg 49:980-987, 2008. 15. Suami H, Taylor GI, Pan WR. A new radiographic cadaver injection technique for investigating the lymphatic system. Plast Reconstr Surg 115:2007-2013, 2005. 16. Pan WR, Suami H, Taylor GI. The lymphatic drainage of the superficial tissues of the head and neck: an anatomical study and clinical implications. Plast Reconstr Surg 121:1614-1624, 2008. 17. Suami H, O’Neill J, Pan WR, et al. Superficial lymphatic system of the upper torso: preliminary radiographic results in human cadavers. Plast Reconstr Surg 121:1231-1239, 2008. 18. Suami H, Pan WR, Mann GB, et al. The lymphatic anatomy of the breast and its implications for sentinel lymph node biopsy: a human cadaver study. Ann Surg Oncol 15:863-871, 2008. 19. Pan WR, Suami H, Corlett RJ, et al. Lymphatic drainage of the nasal fossae and nasopharynx: preliminary anatomical and radiological study with clinical implications. Head Neck 31:52-57, 2009. 20. Pan WR, Rozen WR, Stella D, et al. A three-dimensional analysis of the lymphatics of a bilateral breast specimen: a human cadaveric study. Clin Breast Cancer 9:86-91, 2009. 21. Pan WR, le Roux CM. Blood supply to the lymphatic vessels in the leg: an incidental finding. Clin Anat 23:451-454, 2010. 22. Pan WR, le Roux CM, Levy SM, et al. The morphology of the human lymphatic vessels in the head and neck. Clin Anat 23:654-661, 2010. 23. Pan WR, le Roux CM, Levy SM, et al. Lymphatic drainage of the tongue and the soft palate: anatomic study and clinical implications. Eur J Plast Surg 33:251-257, 2010. 24. Pan WR, le Roux CM, Levy SM. Alternative lymphatic drainage routes from the lateral heel to the inguinal lymph nodes: anatomic study and clinical implications. ANZ J Surg 81:431-435, 2010. 25. Pan WR, le Roux CM, Levy SM, et al. Lymphatic drainage of the external ear. Head Neck 33:60-64, 2011. 26. Pan WR, le Roux CM, Briggs CA. Variations in the lymphatic drainage pattern of the head and neck: further anatomic studies and clinical implications. Plast Reconstr Surg 127:611-620, 2011. 27. Pan WR, le Roux CM, Briggs CA. Reply: Acute lymphedema of the eyelid after major reconstruction of the medial canthus: the role of the lymphatic drainage pattern. Plast Reconstr Surg 128:372e, 2011. 28. Pan WR, Wang DG, Levy SM, et al. Superficial lymphatic drainage of the lower extremity: anatomic study and clinical implications. Plast Reconstr Surg 132:696-707, 2013. 29. Wallace S, Jackson L, Dodd GD, et al. Lymphatic dynamics in certain abnormal states. Am J Roentgenol Radium Ther Nuc Med 91:1199-1200, 1964. 30. Baker TJ, Gordon HL, Molienko P. Rhytidectomy. Plast Reconstr Surg 59:24-30, 1977. 31. Baker TJ, Gordon HL. Complications of rhytidectomy. Plast Reconstr Surg 40:31-39, 1967. 32. Guy CL, Converse JM, Morello DC. Aesthetic surgery for the aging face. In Converse JM, McCarthy JG, Littler JW, eds. Reconstructive Plastic Surgery, ed 2. Philadelphia: WB Saunders, 1977. 33. Chalasani R, McNab A. Chronic lymphedema of the eyelid: case series. Orbit 29:222-226, 2010. 34. Klapper SR, Patrinely JR. Management of cosmetic eyelid surgery complications. Semin Plast Surg 21:80-93, 2007. 35. Aveta A, Tenna S, Segreto F, et al. Acute lymphedema of the eyelid after major reconstruction of the medial canthus: the role of the lymphatic drainage pattern. Plast Reconstr Surg 128:370e-372e, 2011. 36. Langer I, Guller U, Berclaz G, et al. Morbidity of sentinel lymph node biopsy (SLN) alone versus SLN and completion axillary lymph node dissection after breast cancer surgery: a prospective Swiss multicenter study on 659 patients. Ann Surg 245:452-461, 2007.

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 95

5/27/2015 9:12:20 AM

96

Part II  Anatomy, Physiology, and Lymphangiogenesis

37. Lucci A, McCall LM, Beitsch PD, et al. Surgical complications associated with sentinel lymph node dissection (SLND) plus axillary lymph node dissection compared with SLND alone in the American College of Surgeons Oncology Group Trial Z0011. J Clin Oncol 25:3657-3663, 2007. 38. Suami H, Chang DW. Overview of surgical treatments for breast cancer-related lymphedema. Plastic Reconstr Surg 126:1853-1863, 2010. 39. Viamonte MJ, Rüttimann A. Atlas of Lymphography. New York: Thieme-Stuttgart, 1980. 40. Kubik S, Manestar M. Topographic relationship of the ventromedial lymphatic bundle and the superficial inguinal nodes to the subcutaneous veins. Clin Anat 8:25-28, 1995. 41. Williams AF, Franks PJ, Moffatt CJ. Lymphoedema: estimating the size of the problem. Palliat Med 19:300-313, 2005. 42. O’Brien BM, Mellow CG, Khazanchi RK, et al. Long-term results after microlymphaticovenous anastomoses for the treatment of obstructive lymphedema. Plast Reconstr Surg 85:562-572, 1990. 43. Koshima I, Nanba Y, Tsutsui T, et al. Long-term follow-up after lymphaticovenular anastomosis for lymphedema in the leg. J Reconstr Microsurg 19:209-215, 2003. 44. Takeishi M, Kojima M, Mori K, et al. Primary intrapelvic lymphaticovenular anastomosis following lymph node dissection. Ann Plast Surg 57:300-304, 2006. 45. Demirtas Y, Ozturk N, Yapici O, et al. Supermicrosurgical lymphaticovenular anastomosis and lymphaticovenous implantation for treatment of unilateral lower extremity lymphedema. Microsurgery 29:609-618, 2009. 46. Narushima M, Mihara M, Yamamoto Y, et al. The intravascular stenting method for treatment of extremity lymphedema with multiconfiguration lymphaticovenous anastomoses. Plast Reconstr Surg 125:935-943, 2010. 47. Huang GK, Hu RQ, Liu ZZ, et al. Microlymphaticovenous anastomosis in the treatment of lower limb obstructive lymphedema: analysis of 91 cases. Plast Reconstr Surg 76:671-685, 1985. 48. Baumeister RG, Siuda S. Treatment of lymphedemas by microsurgical lymphatic grafting: what is proved? Plast Reconstr Surg 85:64-74, 1990. 49. Weiss M, Baumeister RG, Hahn K. Dynamic lymph flow imaging in patients with oedema of the lower limb for evaluation of the functional outcome after autologous lymph vessel transplantation: an 8-year follow-up study. Eur Nucl Med Mol Imaging 30:202-206, 2003. 50. Taylor GI, Razaboni RM. Michel Salmon Anatomic Studies: Arteries of the Muscles of the Extremities and the Trunk—Arterial Anastomotic Pathways of the Extremities. St Louis: Quality Medical Publishing, 1994. 51. Pan WR, Taylor GI. The angiosomes of the thigh and buttock. Plast Reconstr Surg 123:236-249, 2009. 52. Mathes SJ, Nahai F. Reconstructive Surgery: Principles, Anatomy & Technique. St Louis: Quality Medical Publishing, 1997. 53. Strauch B, Yu HL. Atlas of Microvascular Surgery: Anatomy and Operative Approaches, ed 2. New York: Thieme Medical, 2006. 54. Becker C, Hidden G. [Transfer of free lymphatic flaps: Microsurgery and anatomical study] J Mal Vasc 13:119-122, 1988. 55. Gillies HD. Treatment of lymphoedema by plastic operation. Br Med J 1:96-98, 1935. 56. Thompson N. Buried dermal flap operation for chronic lymphoedema of the extremities: ten-year survey of results on 79 cases. Plast Reconstr Surg 45:541-548, 1970. 57. Salvin SA, Upton J, Kaplan WD, et al. An investigation of lymphatic function following free-tissue transfer. Plast Reconstr Surg 99:730-741; discussion 742-743, 1997. 58. Tanaka Y, Tajima S, Imai K, et al. Experience of a new surgical procedure for the treatment of unilateral obstructive lymphedema of the lower extremity: adipo-lymphatico venous transfer. Microsurgery 17:209-216, 1996. 59. Reynolds HM, Dunbar PR, Uren RF, et al. Three-dimensional visualisation of lymphatic drainage patterns in patients with cutaneous melanoma. Lancet Oncol 8:806-812, 2007.

K23349_Neligan_05_Changing Concepts_r4_dc_0061-0096.indd 96

5/27/2015 9:12:20 AM

C hapter 6 Applied Anatomy Miguel Amore, Lucia Tapia, Gisela Romina Pattarone, Diego Mercado

K ey P oints • The lymphatic system plays an important role in transporting tissue fluids and extravasated plasma proteins back to the bloodstream and absorbing lipids from the intestines. • The lymphatic system is composed of collecting vesels and lymph nodes and comprises five distributions: superficial, deep, communicating, perforating, and visceral. • The superficial lymphatic system includes two subsystems: epifascial and interfascial.

App

Research into the anatomy of the lymphatic system and its links to functional status has been and continues to be a controversial subject, in part because of the great complexity of the methods for its visualization and the complexity and variability of the lymphatics between individuals. More than 30 years ago, Caplan and Ciucci of Buenos Aires University began working in the area of vascular anatomy, with a focus on the lymphatic anatomy, developing and adapting a range of techniques for visualization. Our research, the results of which are indicated in this chapter, is a continuation of their work and was carried out by our group in the Third Normal Anatomy Department of Buenos Aires University. In this chapter we will summarize our findings on the anatomy of the lymphatic system, focusing on the lymphatic drainage of the breasts and limbs, to increase understanding of the anatomic basis of lymphedema.

97

K23346_Neligan_06_Applied Anat_r4_0097-0112.indd 97

5/26/2015 8:46:56 AM

98

Part II  Anatomy, Physiology, and Lymphangiogenesis

Research Methods We began our anatomic study of the lymphatic system using a range of staining techniques (such as China ink, latex, Berlin blue, and others) that progressively improved over the years. We primarily used the Gerota method, which employs a mixture of turpentine essence, Prussian blue, and sulfur ether, as described by Dimitrie Gerota in 1896. We modified his method by replacing the sulfur ether with heat to dilate the lymph capillaries. To visualize the lymphatic vessels and the lymph nodes without risk of injuring them during dissection, and to enable our research into the morphology of the lymphatic system, its variant drainage, and the relationship with subfascial lymphatics, we added diaphonization by the Spalteholz technique. This technique is employed after fixation and clearing of the cadaveric material. First the specimen is dehydrated using alcohol, then it is immersed in xylol, thus changing the refractive index and making it possible to generate a three-dimensional image.1,2

General Considerations The lymphatic system plays an important role in transporting tissue fluids and extravasated plasma proteins back to the bloodstream as well as absorbing lipids from the intestinal tract. It is also important for the immune response and is one of the main routes for the metastatic spread of tumor cells. The system starts in the interstitial space with the initial lymphatic capillaries (Fig. 6-1). It drains the lymph through the collecting vessels, which is then filtered in the lymph nodes and finally cleared into the venous system. It is distributed throughout the body, except for the central nervous system, where the perivascular spaces have a prelymphatic or paralymphatic function.

2 1

FIG. 6-1  Imaging of 1, the lymph capillaries and 2, blood vessels.

K23346_Neligan_06_Applied Anat_r4_0097-0112.indd 98

5/26/2015 8:46:57 AM

99

Chapter 6  Applied Anatomy

Lymphatic System Distribution Topographically, the lymphatic system can be separated as follows: • Superficial system: Drains the cutis and subcutis. In terms of the lower-limb lymphatic drainage, in the superficial system there are two subsystems: ȤȤ The epifascial system, between the skin and the saphenous fascia ȤȤ The interfascial system, between the saphenous fascia and the muscle fascia • Deep system: Subaponeurotic/subfascial; drains the lymph from the muscles, joints, synovial sheaths, and nerves. It runs alongside the deep blood vessels. • Communicating system: Consists of a group of lymphatic vessels that interconnect a system (deep or superficial) on the same aponeurotic stratum. • Perforating system: Interconnects the two systems (superficial and deep). • Visceral system: Drains the lymph from different organs.

Lymph Trunks Classically and schematically, the thoracic duct is considered the main prevertebral collector that drains lymph from three body quadrants and discharges it into the left venous angle. It is formed by the union of the two lumbar trunks and the intestinal trunk, at the height of lumbar vertebrae L1 and L2 (abdominal origin) or behind the crus of the diaphragm and in front of thoracic vertebrae T11 and T12 (thoracic origin) (Fig. 6-2).

5 4

6 3

2 1

FIG. 6-2  1, Lumbar trunks; 2, cisterna chyli; 3, azygos vein; 4, superior vena cava; 5, cervical portion of the thoracic duct; 6, esophagus.

K23346_Neligan_06_Applied Anat_r4_0097-0112.indd 99

5/26/2015 8:46:57 AM

100

Part II  Anatomy, Physiology, and Lymphangiogenesis

Occasionally a dilation of the origin can be observed, called the cisterna chyli. Anatomic variations are also frequently seen, such as the plexus shape in the origin and trunk (Fig. 6-3). The thoracic duct ascends between the azygos vein and the aorta and is covered by the pleura and esophagus. At the level of thoracic vertebrae T4 and T5, the duct crosses under the esophagus and aortic arch and ascends toward the thoracic inlet on the left margin of the vertebral column, between the esophagus and the left subclavian artery, reaching the left jugular subclavian angle (Fig. 6-4). Our research has shown that in the left carotid region there is a lymph collector that receives the lymph from the left fascial, cervical, carotid, and temporal regions. The right duct is the one that drains the remaining upper right quadrant to the right venous angle.3,4

A

B 3

2

2

3

1

1

FIG. 6-3  A, 1, Thoracic duct; 2, trachea; 3, esophagus. B, 1, Thoracic duct formation; 2, azygos vein; 3, thoracic aorta artery.

8

6

2

4 3

7

1

FIG. 6-4  1, Jugular-subclavian angle; 2, thoracic duct; 3, left common carotid artery; 4, brachiocephalic arterial trunk; 5, superior vena cava; 6, cervical trachea; 7, left vagus nerve; 8, lymph collector.

5

K23346_Neligan_06_Applied Anat_r4_0097-0112.indd 100

5/26/2015 8:46:58 AM

101

Chapter 6  Applied Anatomy

Applied Anatomy of the Lymphatic System Lymphatic Drainage of the Breast Area The predominant lymphatic drainage pathway from the breast flows toward the axilla (Fig. 6-5). Axillary node dissection is a standard surgical treatment in patients with involved axillary lymph nodes. Unfortunately, arm lymphedema develops in 7% to 77% of patients with axillary lymph node dissections. Sentinel lymph node biopsy has become a frequently used and widely accepted method for surgical staging of axillary lymph nodes in breast cancer, although the incidence of arm lymphedema after sentinel lymph node biopsy varies from 0% to 13%.5 The reasons for this may be seen in the multiple interpretations of the term sentinel lymph node—variously identified as the first lymph node to which the tumor drains, the closest node to the tumor, or the node that is most seen with the probe (which answers only the first word of the term). Thus, under certain circumstances, when a biopsy of more than one node is performed, a minimally invasive surgery becomes a pseudoaxillary lymph node dissection procedure.6 In our anatomic research, we divided the lymphatic drainage of the mammary gland from that of the mammary skin and found it difficult to extrapolate our research to the clinical and surgical practice (Fig. 6-6). The most representative instance was the injection method in the sentinel

FIG. 6-5  1, Breast area; 2, axillary lymph nodes; 3, superficial lymphatic network.

2

1 3

3

FIG. 6-6  1, Breast area; 2, cutaneous lymphatic network; 3, axillary lymph nodes.

2

1

K23346_Neligan_06_Applied Anat_r4_0097-0112.indd 101

5/26/2015 8:46:59 AM

102

Part II  Anatomy, Physiology, and Lymphangiogenesis

lymph node technique. The optimal sites of dye and/or colloid injection have not been defined: intradermic, subareolar, peritumoral, or intratumoral. There is no consensus on this topic. The problem is to consider the same lymphatic drainage of the mammary gland and the mammary skin as a unique functional unit based on the embryologic hypothesis. Under this concept, the subareolar plexus is considered the center of lymphatic drainage in the breast. This plexus was described by Sappey in 1874. Perhaps, as suggested by Turner Warwick, Sappey confused the milk ducts with lymph vessels. He emphasized the subareolar plexus considering that this technique is the mercury injection in cadaver and it dates from 1800.7,8 According to this concept, the exact tumor site is not as relevant at the moment of injection, because the tumor could be located in any of the breast quadrants. However, some investigators advocate injection in the subareolar area. The lymph produced in the mammary parenchyma goes through a perilobular lymphatic network and through the interlobular spaces that feed the lymph capillaries, which meet and lead to the secondary pedicles. These lymph vessels exit the mammary gland and make up the axillary, mediastinal, and retropectoral lymphatic pedicles.9-11

Axillary Pedicle The axillary pedicle is the largest of the three mammary lymphatic pedicles; it is formed by two to six lymphatic vessels. This pedicle may receive lymph vessels from any breast quadrant, either superficial or deep, from the nipple, areola, or the skin covering the breast. It emerges from the lateral region of the breast, following the border of the pectoralis major, passes to the base of the axilla, and then crosses the pectoroaxillary aponeurosis (Fig. 6-7). Four pedicles secondary to the axillary pedicle can be identified: 1. Lateral mammary: The lateral mammary pedicle follows the path taken by the lateral mammary vessels. It is formed by two to five lymphatic vessels and joins the nodes in the lateral mammary chain. This is the first node level of the axilla.

2

3

1

FIG. 6-7  1, Breast area; 2, axillary lymph nodes; 3, axillary lymphatic pedicle. Inset: Closeup view.

K23346_Neligan_06_Applied Anat_r4_0097-0112.indd 102

5/26/2015 8:46:59 AM

103

Chapter 6  Applied Anatomy





2. Subscapular: The subscapular pedicle is little developed. It passes toward the posterior surface of the axilla, where the subscapular vessels lie. It consists of one or two lymphatic vessels and joins the nodes in the subscapular chain. 3. Superior thoracic: The superior thoracic pedicle is formed by a single lymphatic vessel that crosses the aponeurotic base of the axilla, passes toward the apex of the axilla, and then slips in front of the anterior serratus and behind the pectoral muscles, to end in the superior thoracic chain. It is the second node level of the axilla. 4. Axillary-vein: The axillary-vein pedicle occurs least frequently. It is so named because it passes directly into the highest part of the axillary space and lies in direct contact with the axillary vein.

Mediastinal Pedicle The mediastinal pedicle emerges from the medial part of the mammary gland, either in the superficial plane or at a deeper level, and reaches the anterior surface of the fascia of the pectoralis major by following, in some cases, the perforating branches of the internal mammary vessels. During its passage, it is associated with two secondary pedicles that because of their relationship with the pectoralis major muscle, have been named the prepectoral pedicle and subpectoral pedicle (Fig. 6-8). Two pedicles secondary to the mediastinal pedicle have been identified: 1. Prepectoral: Some lymphatic vessels run across the anterior surface of the pectoralis major muscle before penetrating between the fibers of the costosternal part of this muscle, either in the middle of the muscle or close to its sternal border; others can cross the pectoralis major immediately and pass between it and the intercostal muscles toward the nodes of the internal mammary chain. 2. Subpectoral: As it leaves the mammary gland, the subpectoral pedicle passes downward toward the inferolateral border of the pectoralis major muscle and then rapidly insinuates itself between this muscle and the superior attachment of the rectus abdominis muscle.

2

1 3 3

FIG. 6-8  Inner view of the thoracic wall. 1, Breast area; 2, internal mammary chain; 3, perforating lymph vessels.

K23346_Neligan_06_Applied Anat_r4_0097-0112.indd 103

5/26/2015 8:47:00 AM

104

Part II  Anatomy, Physiology, and Lymphangiogenesis

Retropectoral Pedicle The retropectoral pedicle emerges from the posterior surface of the mammary gland and passes toward the pectoralis major muscle, into which it enters together with the thoracoacromial (acromiothoracic) vessels. When it passes through the costosternal part of the pectoralis major muscle, it is possible to identify three secondary pedicles: transpectoral, interpectoral, and pectoroaxillary. Three pedicles secondary to the retropectoral pedicle have been identified: 1. Transpectoral: The transpectoral pedicle owes its name to the fact that the lymphatic vessels pass through both pectoral muscles. It is formed by a single lymphatic vessel that drains directly into the superior thoracic chain or into the axillary chain. 2. Interpectoral (Grosmann-Rotter): The interpectoral pedicle is the most frequently found of the posterior pedicles. It is formed by one or two lymphatic vessels that perforate the pectoralis major muscle, following the acromiothoracic vessels and draining in the interpectoral chain made up of two to six lymph nodes. 3. Pectoroaxillary: The pectoroaxillary pedicle is formed by a single lymphatic vessel that crosses the pectoralis major and emerges by its posterior surface. It drains in the external mammary chain. It is interesting to analyze the role of the perforating lymph vessels, which are able to communicate a superficial network with the deep lymphatic system. This is observed in some specimens, where there is evidence of the connection between the superficial systems by a perforating lymph vessel to the internal mammary chain.

Lymphatic Drainage of the Upper Limb Superficial Lymphatic System Pathways The superficial lymphatic drainage of the upper limb runs mainly on its anterior surface. At the level of the forearm and upper arm one can find different lymphatic pathways taking the axis of the superficial venous system.9-13 Pathways of the Forearm • Anterior radial: Runs obliquely following the superficial radial vein and it is formed by 3 to 10 lymph vessels. • Posterior radial: Formed between 5 and 15 lymph vessels and helps with the formation of the internal or bicipital and the external or cephalic pathways. • Anterior ulnar: Runs obliquely from the hypothenar region of the hand to the fold of the elbow, from inside to outside, accompanying the superficial ulnar vein. It is formed by 5 to 8 lymphatic vessels. • Posterior ulnar: Found in the posterior face of the wrist and the lower third of the forearm, where it meets with the anterior pathway, thus contributing to the formation of the anterior or bicipital and the internal or basilic terminal lymphatic pathway. It is made up of 5 to 15 lymphatic vessels.

K23346_Neligan_06_Applied Anat_r4_0097-0112.indd 104

5/26/2015 8:47:00 AM

Chapter 6  Applied Anatomy

105

Pathways of the Arm • Bicipital: Formed by 9 to 17 lymphatic vessels. It runs obliquely from the elbow to the base of the axilla, where it goes through the superficial aponeurosis to reach the different node groups of the axilla. • Basilic: Accompanies the basilic vein and is formed by 2 or 3 lymphatic vessels, being the continuation of the anterior and posterior ulnar pathways. It penetrates the deep area of the arm, at different levels, and is continued by the humeral (deep) pathway, thus reaching the deep nodes of the axilla. • Cephalic: Formed by a single vessel, satellite of the cephalic vein, which goes through the external bicipital channel and goes on through the deltopectoral groove, thus ending in the axillar region, or passing over the clavicle and ending in the supraclavicular region. • Posterior: Runs through the posterior-external face of the arm, following the deltotricipital groove. It drains in the scapular circumflex nodes.

Deep Lymphatic System The deep lymphatic system is not described here because its anatomy is beyond the scope of this chapter.

Axillary Lymph Nodes The axillary nodes are the main center of the lymphatic drainage of the upper limb and of the anterolateral and posterolateral regions of the thoracic wall, including the mammary gland. We used the classification proposed by Caplan in 197414 concerning the venous drainage of the axilla. He described four main chains: • Lateral mammary chain: Located on the front of the axillary vessels, in the anterior wall of the axillary space, and runs along the lateral mammary vessels, between the second and seventh rib. This lymph node chain is the most important center of the lymphatic drainage of the upper limb as well as of the breast and skin from the anterior and posterior region of the thorax. Its efferent vessels head to the apex of the axilla to drain their lymph in the upper thoracic chain and the axillary chain, which make up the infraclavicular group described by earlier authors. • Upper thoracic chain: Located in the inner wall of the axilla, behind the pectoral muscles, following the upper thoracic vessels. The efferent lymphatic vessels of this chain drain its lymph in the lower chain of the axillary vein, called the infraclavicular chain by earlier authors. • Subscapular chain: A satellite of the subscapular vessels and a partial satellite of the nerve of the wide dorsal muscle. It receives the drainage of the posterior thoracic wall and, in a small percentage of cases, the lymphatic drainage of the anterior and internal thoracic wall. The efferent pathways of this chain lead to the external mammary gland or the apex of the axilla to drain its lymph in the axillary vein chain. • Axillary vein chain: Located in the upper region of the axillary space, running from the pectoral-axillary base to the apex. This chain is made up of 8 to 10 lymph nodes and four secondary chains: anterior, posterior, upper, and lower. This chain receives the lymphatic drainage from all the regions of the upper limb, from the anterior and posterior thoracic wall, and from the rest of the axillary chains. The efferent pathways generally follow on to the axillary vein to finally reach the venous jugular-subclavial angle or become part of the great lymphatic vein on the right side.

K23346_Neligan_06_Applied Anat_r4_0097-0112.indd 105

5/26/2015 8:47:00 AM

106

Part II  Anatomy, Physiology, and Lymphangiogenesis

2

3

1

FIG. 6-9  Upper limb superficial lymphatic network. 1, Axillary lymph nodes; 2, cephalic derivative pathway; 3, epitrochlear lymph nodes.

Derivative Lymphatic Pathways The derivative lymphatic pathways are lymphatic vessels that do not have a node station in the axilla. These are the cephalic pathway (Mascagni), deltotricipital pathway (Caplan), and the intraaxillary pathway (Ciucci) (Fig. 6-9). Knowledge of these derivative pathways represents one of the anatomic bases for rehabilitative therapy in patients who have upper limb lymphedema, because the pathways are stimulated by the physical therapist through manual lymphatic drainage.

Lymphatic Drainage of the Lower Limb In the lower limb, the lymphatic system can be divided into a superficial and a deep system, taking the muscle fascia–aponeurosis as the axis for this division. Within the superficial system is the epifascial system, between the skin and the saphenous fascia. It is the most developed subsystem in terms of collectors because of the origins of its network of skin sectors: it runs along the width of the subcutaneous cell tissue, accompanying the superficial veins, until it reaches the inguinal lymph nodes. The interfascial system, between the saphenous fascia and the muscle fascia, has few collectors. The deep system, which is smaller, starts in the osteo-articular-muscular region, and its lymphatic collectors accompany it all the way to the vascular packages, to drain themselves in the external iliac chain. Along the way they travel through different lymph node groups in the popliteal region and, less frequently, the nodes of the femoral chain in the thigh. Finally, there are the perforating lymphatic vessels that interconnect both systems.15-17

K23346_Neligan_06_Applied Anat_r4_0097-0112.indd 106

5/26/2015 8:47:00 AM

107

Chapter 6  Applied Anatomy

Superficial Lymphatic System Pathways The superficial vessels start at the foot, where the plantar lymphatic vessels can be distinguished; these are distributed as a network along the foot and are related to the venous sole, as described by Quènu and Lejars. This lymphatic network presents different morphologic features regarding the plantar support points; adopting the plexus feature in the anterior and posterior region, these being the zones with the most pressure.18 This is in contrast to the middle sector, where there are transverse collectors that drain lymph to the posterior of the foot. This lymphatic distribution of the sole of the foot enables us to extrapolate the concept of the superficial venous pump described by Quènu and Lejars to the lymphatic system of this region, which is widely distributed and activated by minimal pressure, draining lymph to the posterior of the foot and contributing to form the pathways of the leg (Figs. 6-10 and 6-11).

2 1

FIG. 6-10  1, Superficial lymphatics; 2, venous network of the sole of the foot.

1

2

FIG. 6-11  Superficial lymphatic network of the lower limb. 1, Inguinal nodes; 2, femoral great saphenous current; 3, tibial great saphenous current. Inset: Closeup view of nodes.

3

K23346_Neligan_06_Applied Anat_r4_0097-0112.indd 107

5/26/2015 8:47:01 AM

108

Part II  Anatomy, Physiology, and Lymphangiogenesis

TABLE 6-1  Pathways of the Superficial Lymphatic System Pathway

Number of Vessels Involved

Location

Forearm Anterior radial

3-10

Runs obliquely following the superficial radial vein

Posterior radial

5-15

Helps with the formation of the internal or bicipital and the external or cephalic pathways

Anterior ulnar

5-8

Runs obliquely from the hypothenar region of the hand to the fold of the elbow, from inside to outside, accompanying the superficial ulnar vein

Posterior ulnar

5-15

Found in the posterior face of the wrist and the lower third of the forearm, where it meets with the anterior pathway, thus contributing to the formation of the anterior or bicipital and the internal or basilic terminal lymphatic pathway

Bicipital

9-17

Runs obliquely from the elbow to the base of the axilla, where it goes through the superficial aponeurosis to reach the different node groups of the axilla

Basilic

2 or 3

Accompanies the basilic vein; the basilic vessel is the continuation of the anterior and posterior ulnar pathways; it penetrates the deep area of the arm at different levels and is continued by the humeral (deep) pathway, thus reaching the deep nodes of the axilla

Arm

Cephalic

1

Posterior

Satellite of the cephalic vein, which goes through the external bicipital channel and on through the deltopectoral groove, thus ending in the axillary region, or passing over the clavicle and ending in the supraclavicular region Runs through the posterior external face of the arm, following the deltotricipital groove; it drains in the scapular circumflex nodes

At the level of the leg and thigh, with the great saphenous vein as the axis, different lymphatic paths can be identified (Table 6-1).

Deep Lymphatic System The few vessels of the deep lymphatic system are part of the deep vascular package and are not described here, because they are beyond the scope of this chapter.

Inguinal Nodes The inguinal nodes form the main center of lymphatic drainage for the lower limb. There are 7 to 17 nodes, all located at the superficial fascia. It is a mistake to include deep inguinal nodes in this classification. It is also inexact to include the classic Cloquet, Rosenmüller, or Pirogoff node within the crural ring or channel as a member of the upper deep inguinal group, because it belongs to the medial group of the external iliac chain.

K23346_Neligan_06_Applied Anat_r4_0097-0112.indd 108

5/26/2015 8:47:01 AM

Chapter 6  Applied Anatomy

109

The nodes located in the internal face of the femoral vein, between the internal lower edge of the crural ring and the upper edge of the arch of the great saphenous vein, are not deep nodes, because at that level the superficial aponeurosis is bound to the pectineal aponeurosis. The nodes located in the medial region of the femoral vein, under the saphenous arch, should not be considered deep inguinal nodes, but nodes belonging to the femoral vessel chain. From the thirteenth century to date, a number of authors have described the inguinal nodes and have proposed different nomenclatures to classify the existing node groups,3,19-22 but the one that has stood the test of time is the one proposed by Quènu and Lejars,23 who classified the groups using two imaginary lines, one perpendicular and one vertical, in relation to the axis of the great saphenous vein. This classification allows these nodes to be grouped in four groups and a central one. In our research we were able to observe that there is a close relation between these nodes and the ones that branch from the great saphenous vein in the femoral saphenous junction. The lymph node vessels from this region drain in the various tributaries of the saphenous vein or in the vein itself. It is of utmost importance for anatomic repair of this vein and its tributaries to classify the inguinal nodes. Surgeons must take into account the random anatomic variations of the superficial venous system to avoid injuries to the lymphatic vessels. Optimal anatomic knowledge is essential to safe and successful surgery in this region. With these criteria, we can distinguish the following veins as tributary veins of the great saphenous vein: • Circumflex iliac vein • Superficial epigastric vein • Pudendal vein • Anterior accessory of the great saphenous vein Thus five very different groups can be defined: three upper groups and two lower ones (Figs. 6-12 and 6-13). • Circumflex iliac group: Located below the external third of the crural arch and closely related to the iliac circumflex vein. This group is constant and varies from 2 to 5 nodes. It has afferent vessels from the cutaneous area of the lumbar, gluteal, and external abdominal regions, the external genitals, the anal canal, and the lower limb. It has efferent vessels that are related to the superficial epigastric group and the external iliac chain. • Superficial epigastric group: Located below the middle third of the crural arch and related to the superficial epigastric vein. It is constant and varies from 2 to 3 nodes. It receives afferent vessels from the cutaneous area of the anterior abdominal and lumbar regions, the external genitals, and the anal canal. Its efferent vessels flow to the pudendal group and the iliac chain. • Pudendal group: Located below the internal third of the crural arch and above the pectineus muscle. This group has an intrinsic relation with the external pudendal veins, is constant, and has from 2 to 4 nodes. It receives afferent vessels from the external genital, anal, and abdominal regions, and a smaller proportion from the lower limb. Its efferent vessels flow to the crural ring to reach the external iliac chain, and it is the main receptor of the Cloquet, Rosemüller, or Pirogoff node, mistakenly considered a deep inguinal node.

K23346_Neligan_06_Applied Anat_r4_0097-0112.indd 109

5/26/2015 8:47:01 AM

110

Part II  Anatomy, Physiology, and Lymphangiogenesis

2

1

FIG. 6-12  1, Inguinal lymph nodes; 2, iliac lymph nodes. Inset: Closeup view of nodes.

3 2

FIG. 6-13  1, Deep lymph vessels; 2, inguinal lymph nodes; 3, iliac lymph nodes. 1

• Medial saphenous group: Located below the falciform ligament, inside the great saphenous vein. This group is constant and is made up of 1 to 3 nodes, of which 1 is frequently found. It receives afferent vessels from the lower limb, the external genitals, the anal canal, and the cutaneous area of the perineum. Its efferent vessels flow to the upper groups and the external iliac chain. • Lateral saphenous group: Located below the lower part of the falciform ligament and outside the anterior accessory vein. It is constant and frequently consists of 1 node. It mainly receives afferent vessels from the lower limb. Its efferent vessels flow to the upper groups, to the medial great saphenous vein, and to the external iliac chain. The difference in the afferent vessels between the upper groups in comparison with the lower groups means that it is of the utmost importance to preserve, to the degree possible, the lower groups—the ones that receive the greatest afferent vessels from the lower limb. Surgical injuries of

K23346_Neligan_06_Applied Anat_r4_0097-0112.indd 110

5/26/2015 8:47:02 AM

Chapter 6  Applied Anatomy

111

the lymphatic system can occur during even a minimal intervention in this area, such as a simple biopsy, or as an unnoticed injury caused during venous surgery. This is the reason that in lymph node transfers, external upper inguinal nodes are frequently used to preserve the lower groups. Surgeons should not only be cautious of causing secondary injuries in the inguinal region, but also those that might occur in the trunk of the great saphenous vein, considering the relationship with the superficial lymphatic system in the internal leg. The distribution of the superficial lymphatic system in the lower limb is located mainly on the epifascial stratus and not on the interfascial one, together with the great saphenous vein. Saphenous stripping may have increased the likelihood of secondary injuries of the lymphatic system; today this is much less likely. Laser techniques and radiofrequency procedures are usually the best choice to completely avoid secondary injuries. However, if these technologies are used incorrectly, their thermal radiation may injure surrounding tissues.

Superficial Popliteal Node Several authors, such as Mascagni, Bardeleben, Haekel and Froshe, Poirier and Cuneo, Ehinger, Baum, and Jdanov, among others, who have observed this node. It is important to take this node into account when performing lymphoscintigraphy, because visualization of the nodes at the popliteal level is a marker of hypertension in the superficial system, referring to deep nodes and marking the difference with the superficial popliteal node, which is a simple node station belonging to the frequent solitaire nodes. It is located in the region of the popliteal fossa and is closely related to the small saphenous vein. It is represented by a single node and receives afferent vessels from the surrounding sectors and their efferent vessels drain to the deep popliteal nodes.

R EFERENCES 1. Houssay B. La Investigación Científica. Buenos Aires, Argentina: Ed Columba, 1960. 2. Pissas A. Dissertation à propos des téchniques d’injection cadaverique des vaisseaux lymphatiques et leur contribution à la connaissance du drainage lymphatique des viscères. Memoire. Presente a l’Univiersité Paris Nord, Paris, 1979. 3. Amore M, Verges J. Variación de la técnica de diafanización aplicada al sistema linfático. Presented at the Thirty-sixth Congreso Rioplatense de Anatomía, Mendoza, Argentina, 1999. 4. Bartels P. Das Lymphgefabsystem. In Bardeleben’s Handbuch der Anatomie des Menschen. Jena, Germany: G Fisher, 1909. 5. Poirier P, Cuneo B. Étude spéciale des lymphatiques des differentes parties du corp. In Poirier P, Charpy A. Traité de l’Anatomie Humaine. Part II, vol 4. Paris: Masson, 1902. 6. Rovuiere H. Anatomie des Lymphatiques de l’Homme. Paris: Masson, 1932. 7. Földi M, Földi E, Kubik S. Textbook of Lymphology. Munich: Elsevier, 2003. 8. Kiefer F, Schulte-Marker S. Developmental aspect of the lymphatic vascular system. Vienna: SpringerVerlag, 2014. 9. Dieter R, Dieter R Jr, Dieter R III. Venous and lymphatic diseases. New York: McGraw-Hill, 2011. 10. Campisi CC, Amore M. Lymphatic drainage of mammary gland: from anatomy to surgery to microsurgery. Progress in Lymphology XXIII. 45(Suppl):75-78, 2012. 11. Amore M. Review the lymphatic anatomy on the sentinel node era. Progress in Lymphology XXIII. 45(Suppl):99, 2012.

K23346_Neligan_06_Applied Anat_r4_0097-0112.indd 111

5/26/2015 8:47:03 AM

112

Part II  Anatomy, Physiology, and Lymphangiogenesis

12. Suami H, Pan WR, Mann GB, Taylor GI. The lymphatic anatomy of the breast and its implications for sentinel lymph node biopsy: a human cadaver study. Ann Surg Oncol 15:863-871, 2008. 13. Borgstein PJ, Meijer S, Pijpers RJ, et al. Functional lymphatic anatomy for sentinel node biopsy in breast cancer: echoes from the past and the periareolar blue method. Ann Surg 232:81-89, 2000. 14. Etude des Centres Lymphoganglionnaires superficiels et profonds du membre supérieur. 58 Congrès de l’Asociation des Anatomistes. Liège, 1974. 15. Caplan I. Lymphatic drainage of the mammary gland. Ann Clin 4:329-335, 1982. 16. Caplan I. Estudio e Investigación para una División de la Distribución Linfática Ganglionar de la Región Axilar dada por la Estrella Venosa de la Misma. Buenos Aires, Argentina: La Semana Medica, 1958. 17. Ciucci JL. Linfedema del Miembro Superior. Buenos Aires, Argentina: Nayarit, 2009. 18. Caplan I. Le système lymphatique du pouce. Mémoires du Laboratoire d’Anatomie de la Faculté de Médecine de Paris. 32:5-65, 1977 19. Ciucci JL. Doctoral thesis. Grandes corrientes linfáticas del miembro superior. Buenos Aires University, Buenos Aires, Argentina, 1987. 20. Caplan I. Sugestiones para una Nueva División Topográfica de la Distribución Linfática de la Región Inguinofemoral, dada por la Estrella Venosa de Scarpa. Buenos Aires, Argentina: La Prensa Medica, 1957. 21. Caplan I. Doctoral thesis. El sistema linfático ganglionar de la región poplítea. Buenos Aires University, Buenos Aires, Argentina, 1966. 22. Ciucci JL. Linfedema de los Miembros Inferiores. Buenos Aires, Argentina: Nayarit, 2009. 23. Quènu E, Lejars F. Les Veines de la Plante du Pied Chez l’Homme et le Grands Animaux. Ètudes sur le Système Circulatoire, vol I. Paris: G Steinheil, 1894.

K23346_Neligan_06_Applied Anat_r4_0097-0112.indd 112

5/26/2015 8:47:03 AM

C hapter 7 Lymphangiogenesis Jeremy S. Torrisi, Ira L. Savetsky, Jason C. Gardenier, Babak J. Mehrara

K ey P oints • Lymphangiogenesis is the formation of new lymphatic vessels. • Lymphangiogenesis is regulated by both prolymphangiogenic and antilymphangiogenic mechanisms. • B cells and macrophages are key sources of prolymphangiogenic growth factors, such as vascular endothelial growth factors.

Lym

• T cells are a key source of antilymphangiogenic cytokines, such as interferon gamma.

Lymphangiogenesis is the formation of new lymphatic vessels. Specifically, lymphangiogenesis refers to the sprouting of lymphatic vessels from preexisting vessels with migration and differentiation of lymphatic endothelial cells (LECs). In contrast, lymphovasculogenesis refers to the de novo formation of lymphatic vessels from mesenchymal-derived sources. These processes are not mutually exclusive and contribute to varying degrees to lymphatic vessel development and proliferation. This process is regulated by a complex interaction between prolymphangiogenic and antilymphangiogenic forces, the balance of which modulates a wide variety of effects in LECs, including migration, differentiation, proliferation, and survival.

Prolymphangiogenic Mechanisms The discovery of lymphatic-specific markers such as Prox1, LYVE1, and podoplanin led to a significant increase in our understanding of the mechanisms that regulate lymphatic development and regeneration. These studies have identified several growth factors and cytokines that either directly or indirectly regulate LEC differentiation, proliferation, migration, and function. These molecules are expressed in various tissues and have overlapping functions. In general, the expression of lymphangiogenic factors leads to lymphatic vessel growth and regeneration, thereby increasing lymphatic function, although there are some tissue and temporal differences in the mechanisms of action. Although initial studies in this field focused on vascular endothelial growth 113

K23346_Neligan_07_Lymphangiogen_r2_dc_0113-0120.indd 113

5/26/2015 8:49:50 AM

114

Part II  Anatomy, Physiology, and Lymphangiogenesis

factors, more recent studies have identified several crucial lymphatic regulators, which suggests that lymphatic homeostasis is a complex process in vivo.

Vascular Endothelial Growth Factors A crucial factor for LEC development is vascular endothelial growth factor C (VEGF-C). VEGF-C interacts with vascular endothelial growth factor receptor 2 (VEGFR-2) and vascular endothelial growth factor receptor 3 (VEGFR-3) and regulates a wide range of effects in LECs, including differentiation, survival, and migration. Vascular endothelial growth factor D (VEGF-D) is a closely related homolog that also activates VEGFR-2/VEGFR-3. VEGFR-3 expression is primarily limited to LECs in postnatal mice and is used as a specific marker to identify lymphatic vasculature.1 VEGF-C, and to a lesser extent VEGF-D, are highly expressed in the regions of the body in which the lymphatics develop in abundance.2 Gradients of VEGF-C regulate the migration of LECs during lymphatic repair after wound healing, and VEGF-C overexpression in the skin of transgenic mice results in lymphatic proliferation and vessel enlargement.3 Similarly, gene therapeutic or recombinant protein delivery of VEGF-C results in lymphangiogenesis caused by increased budding, migration, proliferation, and differentiation of LECs.4 The pioneering work of Karkkainen et al2 showed that VEGF-C is essential for lymphatic development, because VEGF-C–deficient mice do not develop lymphatic vasculature and die in utero. Mice heterozygous for VEGF-C have severely hypoplastic lymphatics with significant lymphatic defects, including lymphedema. Although VEGF-C and VEGF-D appear to have overlapping effects, the independent roles of these growth factors are not completely understood, because congenital VEGF-D deficiency is not lethal. However, exogenous delivery of VEGF-D can rescue VEGF-C knockout mice and restore lymphatic differentiation and development. The key roles for VEGF-C in the regulation of lymphatic development and function are also reflected in VEGFR-3– deficient mice and humans. Although homozygous deficiency of VEGFR-3 results in intrauterine death, mice with a heterozygous inactivating mutation of VEGFR-3 develop chylous ascites and primary lymphedema with severely hypoplastic cutaneous lymphatic vessels.4,5 Heterozygous inactivating mutations of VEGFR-3 have also been clinically identified and are the cause of Milroy disease, an autosomal dominant form of primary lymphedema. The exogenous administration of VEGF-C improves lymphatic function in many models of lymphatic insufficiency and dysfunction. For instance, local delivery of VEGF-C in various animal models of primary or secondary lymphedema, including the rabbit ear, rat hindlimb, and mouse tail models, significantly decreases edema, improves lymphangiogenesis, and decreases the pathologic changes associated with lymphedema.6-8 Similarly, the local delivery of VEGF-C in sheep, pig, and rat lymphadenectomy models enhances lymphatic transport and decreases lymphedema.9 Adenoviral VEGF-C delivery induced growth, remodeling, differentiation, and maturation of lymphatic capillaries after lymph node dissection in mouse and pig models.10-13 However, although these results are exciting, the clinical translation of these findings to patients with iatrogenic lymphedema resulting from cancer treatment has been hampered because VEGF-C is a major regulator of tumor growth and metastasis.14-16 In addition, in some preclinical models, the exogenous administration of even massive doses of VEGF-C has not led to an improvement in lymphedema, which suggests that additional mechanisms are involved.17 Therefore understanding these other pathways (see Antilymphangiogenic Mechanisms in this chapter) is critical if lymphangiogenesis and the restoration of lymphatic flow are the ultimate goals.

K23346_Neligan_07_Lymphangiogen_r2_dc_0113-0120.indd 114

5/26/2015 8:49:50 AM

Chapter 7  Lymphangiogenesis

115

VEGF-A (often simply referred to as VEGF) is a well-known regulator of angiogenesis and ac­ tivates VEGFR-2. However, recent work indicates that VEGF-A, which is also a regulator of lymphangiogenesis, plays a central role in this process in some circumstances. For example, inflammatory lymphangiogenesis in the lymph nodes is considered primarily regulated by in­ creased B-cell expression of VEGF-A. These effects are the result of activation of VEGFR-2 on LECs, which subsequently activate a diverse set of intracellular pathways with significant overlap with VEGF-C/VEGFR-3. Similarly, gene therapeutic approaches with VEGF-A promote lymphatic vessel enlargement and proliferation.18,19 However, in contrast to VEGF-C, VEGF-A administration is also associated with the proliferation of immature, leaky blood vessels. Macrophages are a key cellular source of VEGF-C and VEGF-A during wound repair. Indeed, these cells play a key role in the regulation of lymphangiogenesis in several physiologic settings, including inflammation, tumor-associated lymphangiogenesis, and during wound repair. Bone marrow–derived monocytes are recruited by various inflammatory stimuli and differentiate into macrophages that then secrete inflammatory cytokines and chemokines. In addition, macrophages produce large amounts of VEGF-C, thereby promoting LEC migration and differentiation. A few studies have also suggested that macrophages can transdifferentiate into LECs, thereby directly contributing to and incorporating into newly formed lymphatic vessels. Abnormalities in macrophage function and VEGF-C production have been linked to impaired lymphangiogenesis in diabetic mouse models, suggesting that this pathway plays an important physiologic role. Other sources of VEGF-C include B cells and mast cells. However, the relative contribution of these cell types to lymphangiogenesis during wound repair remains less well understood.20,21

Fibroblast Growth Factors Fibroblast growth factors (FGFs) are a family of growth factors that are involved in lymphangiogenesis, angiogenesis, endocrine signaling pathways, and wound healing.22 Although there are many isoforms of FGFs, the lymphangiogenic roles of FGF-1 and FGF-2 have been the most widely studied. These growth factors regulate lymphangiogenesis by direct (for example, direct effects on LECs) and indirect (by activation of VEGFs) mechanisms. However, the preponderance of evidence suggests that indirect mechanisms are more important in vivo. Several lines of evidence propose that these indirect mechanisms are related to increased expression of VEGF-C, VEGF-C independent activation of VEGFR-3, and upregulation of VEGFR-3 expression.23 Thus, although the exact mechanisms by which FGFs regulate lymphangiogenesis remain unknown, it is likely that these growth factors have key roles in vivo and that the effects of these pathways are dose, site, and situation dependent.24

Hepatocyte Growth Factor Hepatocyte growth factor (HGF) is a growth factor produced by various cells of mesenchymal origin. The growth factor is a heparin-binding glycoprotein whose expression is mediated by the binding of the HGF receptor, a large, membrane-spanning receptor kinase.25 HGF directly promotes the proliferation, migration, and tubule formation of LECs. Overexpression of HGF in transgenic mice significantly increases lymphatic vessel proliferation, and the delivery of exogenous HGF in mouse and rat models of lymphedema ameliorates lymphedema.26,27 Recent epidemiologic studies have also shown that HGF signaling plays a key role clinically in lymphan-

K23346_Neligan_07_Lymphangiogen_r2_dc_0113-0120.indd 115

5/26/2015 8:49:50 AM

116

Part II  Anatomy, Physiology, and Lymphangiogenesis

giogenesis and lymphatic repair, because patients with HGF signaling defects had a significantly increased risk of developing secondary lymphedema.28,29

Insulin-Like Growth Factors Insulin-like growth factor1 (IGF-1) and IGF-2 directly regulate lymphangiogenesis by binding cell surface receptors and multiple IGF-binding proteins.30 In contrast to FGFs, the effects of IGFs on inflammatory lymphangiogenesis appear to be independent of VEGFR-3 activation or expression of VEGF-C, which suggests that IGFs have direct effects on LECs. This family causes promotion of lymphangiogenesis both in vivo and in vitro. With the corneal model of lymphangiogenesis, IGF-1 and IGF-2 induce lymphangiogenesis. Interestingly, this effect was not reduced after VEGFR-3 activity was inhibited, which suggests that IGF’s effect on lymphangiogenesis is not mediated by the VEGF-C/VEGFR-3 pathway. IGF-2 stimulates LEC proliferation and migration in vitro.31

Platelet-Derived Growth Factors Like the IGF family, the platelet-derived growth factor (PDGF) family is large and is composed of five different isoforms (PDGF-AA, PDGF-AB, PDGF-BB, PDGF-CC, and PDGF-DD). These isoforms bind to three different tyrosine kinase receptors (PDGF alpha alpha, PDGF beta beta, and PDGF alpha beta).32 PDGFs have potent lymphangiogenic roles regulating LEC proliferation, survival, and migration as demonstrated by in vivo and in vitro studies. PDGF-induced lymphangiogenesis is not inhibited by VEGF-C antagonists or VEGFR-3 blockade, which suggests that the primary mode of action for PDGFs in lymphangiogenesis is a direct one.33 However, other studies have shown that although PDGF has direct effects on lymphangiogenesis, these effects are synergistic with VEGF-C/VEGFR-3 stimulation.34

Antilymphangiogenic Mechanisms The vast majority of studies in the literature have focused on mechanisms that promote lymphangiogenesis. However, recent studies indicate that this process is regulated on multiple levels and represents a balance between prolymphangiogenic and antilymphangiogenic forces. Thus, in some physiologic settings, even high doses of recombinant lymphangiogenic cytokines are overwhelmed by antilymphangiogenic forces, which result in impaired lymphatic function and defective lymphatic regeneration. Furthermore, the balance between prolymphangiogenic and antilymphangiogenic mechanisms is complex and context dependent such that temporal, spatial, or tissue-dependent factors can tip the balance between these opposing mechanisms. Nevertheless, it is clear that experimental approaches designed to improve lymphatic function must take both pathways into consideration and optimize their balance for best results.

Transforming Growth Factor Beta 1 Transforming growth factor beta 1 (TGF-beta 1) is a growth factor that plays a key regulator role in various cellular processes, including embryogenesis, immune responses, tissue and wound repair, and inflammation.35 TGF-beta 1 acts by signaling through transmembrane receptor serine and threonine kinases and regulates multiple intracellular pathways.36 Recent studies have shown that TGF-beta 1 is necessary for lymphatic development during embryogenesis, but serves as a

K23346_Neligan_07_Lymphangiogen_r2_dc_0113-0120.indd 116

5/26/2015 8:49:50 AM

Chapter 7  Lymphangiogenesis

117

negative regulator of lymphangiogenesis during wound repair and in tumor-associated lymph­ angiogenesis. In these circumstances, TGF-beta 1 directly decreases LEC proliferation and tu­ bule formation and negatively regulates genes necessary for lymphatic differentiation.37,38 TGFbeta 1 expression is potently increased in response to lymphatic injury and in lymphedematous tissues, and inhibition of TGF-beta 1 activity decreases fibrosis, increases lymphangiogenesis, and improves lymphatic function in preclinical mouse models.39 These effects of TGF-beta 1 appear to be independent of VEGF-C/VEGFR-3 signaling, because even high levels of VEGF-C do not overcome the negative effects of TGF-beta 1 on lymphatic function. Collectively, these findings suggest that manipulation of TGF-beta 1 function may be a novel way to improve lymphatic function and regeneration in the experimental treatment of lymphedema.

Interferon Gamma Interferon gamma (IFN-gamma) is a cytokine secreted by leukocytes with critical roles in innate and adaptive immune responses.40,41 More recent studies have shown that IFN-gamma, which is similar to TGF-beta 1, has direct antilymphangiogenic effects and plays an important role in the resolution of inflammatory lymphangiogenesis and in the regulation of lymphatics during wound healing. These results are related to various effects of IFN-gamma on LECs, including increased apoptosis, impaired proliferation, decreased migration, and inhibition of functional differentiation.42 In addition, similar to TGF-beta 1, IFN-gamma is bound by specific receptors expressed on the surface of LECs and directly regulates the expression of lymphatic-specific genes.43 Inhibition of IFN-gamma function markedly increases inflammatory lymphangiogenesis and delays its resolution after the inflammatory stimulus has been removed. A primary source of IFN-gamma in this process is T cells, a finding that has led some investigators to suggest that T cells are antilymphangiogenic. These findings are supported by previous studies showing that depletion of T cells markedly increases lymphatic repair after injury and suggests that inhibition of T-cell responses may represent a novel means by which lymphatic repair can be augmented.44

Endostatin Endostatin is a proteolytic fragment originating from collagen XVIII that inhibits endothelial cell proliferation, angiogenesis, and tumor growth.45 More recent studies have shown that endostatin is a potent physiologic inhibitor of lymphangiogenesis in tumor-associated lymphatics.46 In vitro studies have shown that endostatin markedly inhibits proliferation and tubule formation of LECs. These findings support in vivo studies in mice that have shown inhibition of lymphangiogenesis and lymph node metastases of human oral squamous cell carcinomas.47 These effects are related to the direct effects of endostatin on LECs and the indirect modulation of VEGF-C expression and disruption of LEC interaction with integrin alpha 9, a key extracellular matrix molecule regulating LEC proliferation and survival.48,49

Clinical Implications A clearer understanding of the cellular and molecular mechanisms that regulate lymphangiogenesis has important clinical implications. The most significant aspect of this research is the attempt to shift the balance between prolymphangiogenic and antilymphangiogenic forces to increase lymphangiogenesis. For example, blockade of antilymphangiogenic cytokines in combination with

K23346_Neligan_07_Lymphangiogen_r2_dc_0113-0120.indd 117

5/26/2015 8:49:50 AM

118

Part II  Anatomy, Physiology, and Lymphangiogenesis

the delivery of VEGF-C may not only increase lymphatic regeneration potential but also decrease the dose of VEGF-C that is necessary to achieve this end result. This is important in patients with cancer-related lymphedema, because prolymphangiogenic growth factors such as VEGF-C and VEGF-A regulate tumor growth and metastasis. Thus decreasing the dose of these growth factors has important oncologic implications and may aid in clinical translation of preclinical studies. Alternatively, it may be possible to simply downregulate antilymphangiogenic mechanisms without the additional delivery of VEGF-C to achieve this end result. This approach has been reported in preclinical studies showing that blockade of TGF-beta 1 or inhibition of T-cell inflammatory responses can potently increase lymphangiogenesis independent of concomitant increases in VEGF-A or VEGF-C. A combination of these interventions with surgical treatment of lymphedema may lead to improved lymphangiogenesis and with more predictable outcomes, thereby increasing the efficacy of surgical interventions.

Conclusion Lymphangiogenesis is a complex process guided by various discrete but overlapping mechanisms. Understanding these pathways is critical for the development of novel methods designed to improve lymphangiogenesis and lymphatic function. In addition, understanding the balance between prolymphangiogenic and antilymphangiogenic pathways and the cellular sources of molecules that regulate these processes provides additional insight into viable treatment options that can be combined with surgery to augment lymphatic function.

C linical P earls • The treatment of lymphatic dysfunction may be improved by inducing lymphangiogenesis. • Lymphangiogenesis can be facilitated by the upregulation of prolymphangiogenic growth factors and the inhibition of antilymphangiogenic factors. • Augmentation of lymphangiogenesis along with other treatment modalities (for example, surgery and compression) may be necessary to stabilize the disease and improve patient outcomes.

R EFERENCES 1. Tammela T, Enholm B, Alitalo K, et al. The biology of vascular endothelial growth factors. Cardiovasc Res 65:550-563, 2005. 2. Karkkainen MJ, Haiko P, Sainio K, et al. Vascular endothelial growth factor C is required for sprouting of the first lymphatic vessels from embryonic veins. Nat Immunol 5:74-80, 2004. 3. Jeltsch M, Kaipainen A, Joukov V, et al. Hyperplasia of lymphatic vessels in VEGF-C transgenic mice. Science 276:1423-1425, 1997 4. Karpanen T, Alitalo K. Molecular biology and pathology of lymphangiogenesis. Annu Rev Pathol 3:367-397, 2008.

K23346_Neligan_07_Lymphangiogen_r2_dc_0113-0120.indd 118

5/26/2015 8:49:50 AM

Chapter 7  Lymphangiogenesis

119

5. Alitalo K, Tammela T, Petrova TV. Lymphangiogenesis in development and human disease. Nature 438:946-953, 2005. 6. Szuba A, Skobe M, Karkkainen MJ, et al. Therapeutic lymphangiogenesis with human recombinant VEGF-C. FASEB J 16:1985-1987, 2002. 7. Yoon YS, Murayama T, Gravereaux E, et al. VEGF-C gene therapy augments postnatal lymphangiogenesis and ameliorates secondary lymphedema. J Clin Invest 111:717-725, 2003. 8. Liu Y, Fang Y, Dong P, et al. Effect of vascular endothelial growth factor C (VEGF-C) gene transfer in rat model of secondary lymphedema. Vasc Pharmacol 49:44-50, 2008. 9. Baker A, Kim H, Semple JL, et al. Experimental assessment of pro-lymphangiogenic growth factors in the treatment of post-surgical lymphedema following lymphadenectomy. Breast Cancer Res 12:R70, 2010. 10. Tammela T, Saaristo A, Holopainen T, et al. Therapeutic differentiation and maturation of lymphatic vessels after lymph node dissection and transplantation. Nat Med 13:1458-1466, 2007. 11. Lähteenvuo M, Honkonen K, Tervala T, et al. Growth factor therapy and autologous lymph node transfer in lymphedema. Circulation 123:613-620, 2011. 12. Honkonen KM, Visuri MT, Tervala TV, et al. Lymph node transfer and perinodal lymphatic growth factor treatment for lymphedema. Ann Surg 257:961-967, 2013. 13. Sommer T, Buettner M, Bruns F, et al. Improved regeneration of autologous transplanted lymph node fragments by VEGF-C treatment. Anat Rec (Hoboken) 295:786-791, 2012. 14. Hicklin DJ, Ellis LM. Role of the vascular endothelial growth factor pathway in tumor growth and angiogenesis. J Clin Oncol 23:1011-1027, 2005. 15. Chen JC, Chang YW, Hong CC, et al. The Role of the VEGF-C/VEGFRs axis in tumor progression and therapy. Int J Mol Sci 14:88-107, 2012. 16. Skobe M, Hamberg LM, Hawighorst T, et al. Concurrent induction of lymphangiogenesis, angiogenesis, and macrophage recruitment by vascular endothelial growth factor-C in melanoma. Am J Pathol 159:893-903, 2001. 17. Uzarski J, Drelles MB, Gibbs SE, et al. The resolution of lymphedema by interstitial flow in the mouse tail skin. Am J Physiol Heart Circ Physiol 294:H1326-H1334, 2008. 18. Nagy JA, Vasile E, Feng D, et al. Vascular permeability factor/vascular endothelial growth factor induces lymphangiogenesis as well as angiogenesis. J Exp Med 196:1497-1506, 2002. 19. Kunstfeld R, Hirakawa S, Hong YK, et al. Induction of cutaneous delayed-type hypersensitivity reactions in VEGF-A transgenic mice results in chronic skin inflammation associated with persistent lymphatic hyperplasia. Blood 104:1048-1057, 2004. 20. Kim H, Kataru RP, Koh GY. Regulation and implications of inflammatory lymphangiogenesis. Trends Immunol 33:350-356, 2012. 21. Kataru RP, Lee YG, Koh GY. Interactions of immune cells and lymphatic vessels. Adv Anat Embryol Cell Biol 214:107-118, 2014. 22. Powers CJ, McLeskey SW, Wellstein A. Fibroblast growth factors, their receptors and signaling. Endocr Relat Cancer 7:165-197, 2000. 23. Kubo H, Cao R, Brakenhielm E, et al. Blockade of vascular endothelial growth factor receptor-3 signaling inhibits fibroblast growth factor-2-induced lymphangiogenesis in mouse cornea. Proc Natl Acad Sci U S A 99:8868-8873, 2002. 24. Chang LK, Garcia-Cardeña G, Farnebo F, et al. Dose-dependent response of FGF-2 for lymphangiogenesis. Proc Natl Acad Sci USA 101:11658-11663, 2004. 25. Giordano S, Di Renzo MF, Narsimhan RP, et al. Biosynthesis of the protein encoded by the c-met protooncogene. Oncogene 4:1383-1388, 1989. 26. Kajiya K, Hirakawa S, Ma B, et al. Hepatocyte growth factor promotes lymphatic vessel formation and function. EMBO J 24:2885-2895, 2005. 27. Saito Y, Nakagami H, Morishita R, et al. Transfection of human hepatocyte growth factor gene ameliorates secondary lymphedema via promotion of lymphangiogenesis. Circulation 114:1177-1184, 2006.

K23346_Neligan_07_Lymphangiogen_r2_dc_0113-0120.indd 119

5/26/2015 8:49:50 AM

120

Part II  Anatomy, Physiology, and Lymphangiogenesis

28. Finegold DN, Schacht V, Kimak MA, et al. HGF and MET mutations in primary and secondary lymphedema. Lymphat Res Biol 6:65-68, 2008. 29. Miaskowski C, Dodd M, Paul SM, et al. Lymphatic and angiogenic candidate genes predict the development of secondary lymphedema following breast cancer surgery. PloS One 8:e60164, 2013. 30. Clemmons DR. Insulin-like growth factor binding proteins and their role in controlling IGF actions. Cytokine Growth Factor Rev 8:45-62, 1997. 31. BjÖrndahl M, Cao R, Nissen LJ, et al. Insulin-like growth factors 1 and 2 induce lymphangiogenesis in vivo. Proc Natl Acad Sci USA 102:15593-15598, 2005. 32. Andrae J, Gallini R, Betsholtz C. Role of platelet-derived growth factors in physiology and medicine. Genes Dev 22:1276-1312, 2008. 33. Cao R, Björndahl MA, Religa P, et al. PDGF-BB induces intratumoral lymphangiogenesis and promotes lymphatic metastasis. Cancer Cell 6:333-345, 2004. 34. Onimaru M, Yonemitsu Y, Fujii T, et al. VEGF-C regulates lymphangiogenesis and capillary stability by regulation of PDGF-B. Am J Physiol Heart Circ Physiol 297:H1685-H1696, 2009. 35. Flanders KC, Major CD, Arabshahi A, et al. Interference with transforming growth factor-beta/Smad3 signaling results in accelerated healing of wounds in previously irradiated skin. Am J Pathol 163:22472257, 2003. 36. Massague J, Blain SW, Lo RS. TGF beta signaling in growth control, cancer, and heritable disorders. Cell 103:295-309, 2000. 37. Oka M, Iwata C, Suzuki HI, et al. Inhibition of endogenous TGF-beta signaling enhances lymphangiogenesis. Blood 111:4571-4579, 2008. 38. Clavin NW, Avraham T, Fernandez J, et al. TGF-beta1 is a negative regulator of lymphatic regeneration during wound repair. Am J Physiol Heart Circ Physiol 295:H2113-H2127, 2008. 39. Avraham T, Daluvoy S, Zampell J, et al. Blockade of transforming growth factor-beta1 accelerates lymphatic regeneration during wound repair. Am J Pathol 177:3202-3214, 2010. 40. Dalton DK, Pitts-Meek S, Keshav S, et al. Multiple defects of immune cell function in mice with disrupted interferon-gamma genes. Science 259:1739-1742, 1993. 41. Schoenborn JR, Wilson CB. Regulation of interferon-gamma during innate and adaptive immune responses. Adv Immunol 96:41-101, 2007. 42. Shao X, Liu C. Influence of IFN-alpha and IFN-gamma on lymphangiogenesis. J Interferon Cytokine Res 26:568-574, 2006. 43. Kataru RP, Kim H, Jang C, et al. T lymphocytes negatively regulate lymph node lymphatic vessel formation. Immunity 34:96-107, 2011. 44. Zampell JC, Yan A, Elhadad S, et al. CD4(1) cells regulate fibrosis and lymphangiogenesis in response to lymphatic fluid stasis. PloS One 7:e49940, 2012. 45. O’Reilly MS, Boehm T, Shing Y, et al. Endostatin: an endogenous inhibitor of angiogenesis and tumor growth. Cell 88:277-285, 1997. 46. Dong X, Zhao X, Xiao T, et al. Endostar, a recombined humanized endostatin, inhibits lymphangiogenesis and lymphatic metastasis of Lewis lung carcinoma xenograft in mice. Thorac Cardiovasc Surg 59:133-136, 2011. 47. Fukumoto S, Morifuji M, Katakura Y, et al. Endostatin inhibits lymph node metastasis by a downregulation of the vascular endothelial growth factor C expression in tumor cells. Clin Exp Metastasis 22:31-38, 2005. 48. Brideau G, Mäkinen MJ, Elamaa H, et al. Endostatin overexpression inhibits lymphangiogenesis and lymph node metastasis in mice. Cancer Res 67:11528-11535, 2007. 49. Ou J, Li J, Pan F, et al. Endostatin suppresses colorectal tumor-induced lymphangiogenesis by inhibiting expression of fibronectin extra domain A and integrin alpha9. J Cell Biochem 112:2106-2114, 2011.

K23346_Neligan_07_Lymphangiogen_r2_dc_0113-0120.indd 120

5/26/2015 8:49:50 AM

C hapter 8 Impact of Genetics on Lymphangiogenesis Kristiana Gordon, Pia Ostergaard

K ey P oints • Primary lymphedema is the presenting symptom of numerous distinct conditions; it is genetically heterogeneous. • Several abnormalities within the genes involved in lymphangiogenesis have been reported to cause primary lymphedema.

Imp

• The identification of more pathogenic genes will advance our knowledge of the etiopathogenesis of lymphatic disease. In time, this may allow the development of therapeutic interventions. • Patients presenting with unexplained lymphedema, even in adulthood, are likely to have an underlying genetically determined primary lymphatic abnormality.

It has become clear that primary lymphedema is not one disease but a clinical sign of several distinct clinical entities. Most patients presenting with lymphedema in adulthood are diagnosed with secondary lymphedema, even without a clear underlying cause. It is likely that many of these patients will have an underlying genetically determined primary lymphatic abnormality that the clinician has not considered. The identification of the molecular abnormality for each subtype of primary lymphedema is crucial, because it advances the understanding of the underlying mechanism of the disease. This knowledge will assist with the identification of new therapeutic strategies in the future. Lymphangiogenesis is the term used to describe the growth of new lymphatic vessels. Developmental lymphangiogenesis occurs in the fetus as the lymphatic system develops. However, lymphangiogenesis may also occur postnatally (for example, in association with inflammatory disease or cancer). This chapter discusses the development of the lymphatic system. (Chapter 7 provides additional detail about the clinical consequences of abnormalities in this process.) 121

K23349_Neligan_08_Genetics_r5_cl_0121-0134.indd 121

5/27/2015 1:43:52 PM

Developmental lymphangiogenesis

122

Part II  Anatomy, Physiology, and Lymphangiogenesis

Vein

Primary lymphatic plexus

VEGF-C

Master vasculature H2O Macromolecules cells Anchoring filaments

Buttonlike junctions

Lymph sac

Smooth muscle cells Luminal valve

LEC progenitors? Prox1 Sox18 COUP-TFII

VEGF-C/VEGFR-3 CCBE1

LEC differentiation

Lymphatic capillaries

Nrp2

FOXC2 Integrin-a9 Ephrin-B2

Sprouting

Zipperlike junctions Collecting vessels

Remodeling

FIG. 8-1  Developmental lymphangiogenesis. The main stages of lymphatic development and the characteristics of normal vessels are shown. The key regulators of different lymphangiogenic processes are indicated in blue. Blood and lymphatic vessels and endothelial cells are shown in red and blue, respectively. (CCBE1, Collagen and calcium-binding EGF domain 1; COUP-TFII, COUP transcription factor II; FOXC2, forkhead box C2; LEC, lymphatic endothelial cell; Nrp2, neuropilin 2; Prox1, prospero homeobox; Sox18, SRY (sex determining region Y)-Box18; VEGF-C, vascular endothelial growth factor C; VEGFR-3, vascular endothelial growth factor receptor 3.)

Martinez-Corral and Makinen1 have schematically summarized developmental lymphangiogenesis in Fig. 8-1. Until recently, the lymphatic system has remained largely ignored by the scientific and medical communities. An overview of the current theories and factors reported to be involved in lymphatic development is presented in this chapter. The literature is continuously updated with newly identified molecules thought to play a role1 (see Chapter 7). Research into lymphangiogenesis has recently identified several lymphatic endothelial cell–specific molecular markers and the genes involved in the process of lymphatic development (Fig. 8-2). Abnormalities within several of these genes are now known to cause different primary lymphedema phenotypes in both humans and animal models (see Chapter 46). However, these recent advances merely represent the “tip of the iceberg,” because there is much about lymphangiogenesis that must still be discovered.

The Origin of Lymphatic Endothelial Cells The first lymph sacs develop in human embryos at the age of 6 to 7 weeks’ gestation, almost 1 month after the blood vasculature system begins to develop.2 Animal model research (see Chapter 46) has demonstrated that a subpopulation of blood endothelial cells, which sprout from cardinal and peripheral veins, will differentiate into lymphatic endothelial cells (LECs).3 Lymphatic vessel endothelial hyaluronan receptor 1 (LYVE1) is the earliest known cell marker indicating that LEC competence has been acquired.4 The LECs will then bud off to form primitive lymphatic structures called lymph sacs.5

K23349_Neligan_08_Genetics_r5_cl_0121-0134.indd 122

5/27/2015 1:43:52 PM

Chapter 8  Impact of Genetics on Lymphangiogenesis

123

FIG. 8-2  The various molecular influences under development of the lymphatic system. (Ang1, 2, 3/4, Angiopoietin 1, 2, 3/4; CCBE1, collagen and calcium-binding EGF domain 1; ECM, endothelial cell membrane; FoxC2, forkhead box C2; IFN- g, interferon-gamma; LEC, lymphatic endothelial cell; LYVE-1, lymphatic vessel endothelial hyaluronan receptor 1; NFATct, nuclear factor of activated T cells, cytoplasmic 1; NP-2, NPK1-related protein kinase 2; Prox1, prospero homeobox; Sox18, SRY (sex determining region Y)-Box18; TGF-b, transforming growth factor beta; Tie1,2, tyrosine kinase with immunoglobulin-like and EGF-like domains 1 and 2; VEGF-C, vascular endothelial growth factor C; VEGF-D, vascular endothelial growth factor D; VEGFR-2/3, vascular endothelial growth factor receptor 2/3.)

The differentiation of LECs from blood vascular endothelial cells is controlled by transcription factors, such as prospero homeobox 1 (Prox1), Sox18, and COUP transcription factor 2 (COUPTFII). Prox1 is required for the differentiation of LECs during embryogenesis and for LEC maintenance during adult life.6 LECs that express Prox1 are then able to upregulate other lymphatic endothelial-specific molecules, such as vascular endothelial growth factor receptor 3 (VEGFR-3) and LYVE1.7 Sox18 is expressed in the cardinal vein and regulates Prox1 expression.8 However, the signal that induces Sox18-mediated Prox1 production has not yet been identified. COUP-TFII interacts with Prox1 and induces expression of LEC-specific genes, including Vegfr-3 and neuropilin 2 (Nrp2).9 It also establishes LEC specification by suppressing Notch signaling.10

K23349_Neligan_08_Genetics_r4_dc_0121-0134.indd 123

5/27/2015 9:20:20 AM

124

Part II  Anatomy, Physiology, and Lymphangiogenesis

Venous Sprouting of Lymphatic Endothelial Cells In embryogenesis, individual LECs connect to each other by adherens junctions and will “sprout” into the primitive lymphatic structures, the lymph sacs.11 Animal studies have identified that the vascular endothelial growth factor C (VEGF-C)/VEGFR-3 and collagen and calcium-binding EGF domain 1 (CCBE1) signaling pathways regulate sprouting.12 The ligands vascular endothelial growth factor D (VEGF-C and VEGF-D) bind to and activate the tyrosine kinase receptors VEGFR-2 and VEGFR-3 and their coreceptor Nrp2 on LECs. VEGFR-3 is considered the main VEGF-C receptor for lymphangiogenesis.1 Both VEGF-C and VEGF-D promote migration and proliferation of LECs in vitro and lymphatic vessel hyperplasia in vivo. However, only VEGF-C is needed for embryonic lymphatic development.13,14 The other VEGF-C receptor, VEGFR-2, is thought to be involved in lymphangiogenesis, but this has not yet been proved. CCBE1 is expressed in tissues that are in close proximity to the budding venous-derived LECs. CCBE1 does not appear to have direct lymphangiogenic activity on its own but can enhance the lymphangiogenic effects of VEGF-C by upregulating the levels of it.15 It was recently discovered that CCBE1 promotes proteolytic cleavage of the poorly active 29/31-kDa form of VEGF-C by the ADAMTS3 protease, resulting in the mature 21/23-kDa form of VEGF-C, which subsequently induces increased VEGF-C receptor signaling.16

Lymphatic Sprouting Further development of the lymphatic system occurs by lymphatic vessel sprouting from the primitive lymph sacs mentioned previously. This process is controlled by the VEGF-C ligand and its receptor VEGFR-3. NRP2, Ephrin-B2, and Notch are also required for the regulation of late embryonic and postnatal lymphatic sprouting and remodeling.17 Notch will induce VEGFR-3 expression and thereby increase endothelial cell responsiveness to VEGF-C in early mouse embryos.18

Remodeling of a Lymphatic Vascular System The primitive lymphatic vessels are now required to mature into a functional vascular network with vessel-type features to serve their critical functions. Remodeling processes occur late in embryonic development and the early postnatal period. They lead to the formation of flap valves in lymphatic capillaries, the establishment of precollecting and collecting vessels by way of smooth muscle cell (SMC) recruitment, and the development of luminal valves. Flaplike openings (called primary valves) form between the LECs of lymphatic capillaries to create button-like intercellular junctions that facilitate the entry of interstitial fluid into initial lymphatic vessels.19 Molecular mechanisms controlling this process remain poorly understood, but vascular endothelial (VE)–cadherin is thought to promote junction stability.19 Intraluminal valves within developing collector vessels are formed by the expression of Prox1 and forkhead box (FoxC2) transcription factors by clusters of LECs. It has been proposed that mechanical forces resulting from the flow of lymph may play a role in establishing the locations

K23349_Neligan_08_Genetics_r4_dc_0121-0134.indd 124

5/27/2015 9:20:20 AM

Chapter 8  Impact of Genetics on Lymphangiogenesis

125

of these valves.20 Valve-forming endothelial cells arrange themselves on the vessel wall to form a transverse ridge, which subsequently develops into mature valve leaflets.21 FOXC2 is a significant regulator of lymphatic valve formation. FOXC2 deficiency in humans and animal models results in lymphatic vessel valve dysplasia.22 However, it is apparent that other regulators are involved in lymphatic valve development, such as NFATC1, calcineurin, Ephrin-B2, Ang2, connexin, and integrin alpha9/fibronectin-EIIIA signaling pathways.21 The lymphatic precollectors and collecting lymphatic vessels are covered by SMCs except in the luminal valve areas to assist with the proximal propulsion of lymph.23 The number of perivascular SMCs increases progressively along the lymphatic vascular trunk. Several markers are involved in SMC recruitment, including platelet-derived growth factor B (PDGF-B).22

Clinical Consequences of Abnormalities Within the Lymphangiogenesis Pathway Until recently primary lymphedema received limited interest from both the clinical and academic communities. Until Connell et al24 published their 2010 paper on classification, phenotyping of patients was considered of little benefit. Their paper highlighted the importance of accurate indepth phenotyping and how it can lead to the identification of new causal genes. A revision of the classification pathway was published in 201325 (Fig. 8-3). It highlighted the spectrum of disorders that may present with primary lymphedema and suggested the underlying causal gene. The classification pathway was presented in the form of a color-coded algorithm to illustrate the five main categories of primary lymphedema and the individual subtypes within these categories. The main categories are as follows: 1. Syndromic disorders associated with lymphedema (but where lymphedema is not the predominant feature) (blue) 2. Localized or generalized lymphedema associated with systemic/visceral lymphatic abnormalities (pink) 3. Lymphedema in association with disturbed growth and/or cutaneous/vascular anomalies (yellow) 4. Congenital lymphedema (green) 5. Late-onset primary lymphedema (purple) There will be exceptions and outliers to this classification system, but it has proved a useful clinical and research tool that is still evolving. Rigorous phenotyping in combination with recent advances in genetic analysis, such as next-generation sequencing, will enhance the identification rate of new genes that cause primary lymphedema. Several abnormalities within genes involved in lymphangiogenesis have already been reported to cause primary lymphedema, and they will be discussed. Although we do not yet fully understand all mechanisms involved in lymphangiogenesis, the identification of mutations in VEGFR-3, VEGF-C, CCBE1, SOX18, and FOXC2 in humans with lymphatic insufficiency supports the critical role these genes play. Mendola et al26 recently screened 78 patients for mutations in known primary lymphedema genes and detected mutations in 36% of cases. This supports the hypothesis that other causal genes for primary lymphedema have yet to be identified. Certainly not all cases of primary lymphedema will have a monogenic cause.

K23349_Neligan_08_Genetics_r4_dc_0121-0134.indd 125

5/27/2015 9:20:20 AM

126

Part II  Anatomy, Physiology, and Lymphangiogenesis

START Proteus syndrome AKT1

Unknown syndrome Syndromic

CLOVE/fibroadipose hyperplasia AKT/PIK3/mTOR pathway

Yes

Known syndrome e.g., Noonan and Turner

No

KT/KT-like

Systemic/visceral involvement prenatal or postnatal onset

Parkes-Weber syndrome RASA1

Some segments Yes All segments

No

Combined vascular malformation Yes

Lymphangioma

No

WILD syndrome

Congenital onset: Lymphedema within first year of life

Late onset: Lymphedema after age 1 yr No

Congenital unisegmental edema

Distichiasis

Lower limb 1 genital edema

Lymphedema-distichiasis syndrome FOXC2

Yes

Multiple segments One limb

Bilateral

Unilateral

No

Late-onset unilateral leg lymphedema

FH 2 ve

Unilateral

Lower limbs only

FH 2 ve Milroy disease FLT4(VEGFR-3) Milroy-like Consider KIF11, VEGF-C

Generalized lymphatic dysplasia (GLD)/ Hennekam syndrome Consider CCBE1

Disturbed growth/ cutaneous manifestations/ vascular anomalies

Lymphangiomatosis/ Gorham syndrome

Congenital multisegmental edema without systemic involvement

Multisegmental lymphatic dysplasia with systemic involvement (MLDSI)

Lower limb

FH 1 ve

Lower limb 1 genitalia

Yes

Bilateral FH 1 ve

No

Meige-like Meige Consider GJC2 Late-onset unisegmental lymphedema Late-onset multisegmental lymphedema Lower limbs 6 genitalia Consider GATA2 Four limb Consider GJC2, Turner

FIG. 8-3  Classification pathway for primary lymphedema. Red section indicates gene tests that are available and should be considered for each subgroup. (CCBE1, Collgen and calcium-binding EGF domain 1; FH, family history; 1ve, positive; 2ve, negative; FOXC2, forkhead box C2; GATA2, GATA binding protein 2; GJC2, gap junction protein, gamma 2; KT/KT-like, KlippelTrenaunay/Klippel-Trenaunay-like; VEGF-C, vascular endothelial growth factor C; VEGFR, vascular endothelial growth factor receptor.)

K23349_Neligan_08_Genetics_r4_dc_0121-0134.indd 126

5/27/2015 9:20:20 AM

Chapter 8  Impact of Genetics on Lymphangiogenesis

127

TABLE 8-1  Pathway for Primary Lymphedema Cutaneous manifestations

Nevi/pigmentation variations (for example, epidermal nevi/vascular malformations)

Distichiasis

Presence of aberrant eyelashes arising from the meibomian glands

Disturbed growth

Hypertrophy (overgrowth) and hypotrophy of bone or soft tissue resulting in altered length of a limb or body part

KT/KT-like

Klippel-Trenaunay/Klippel-Trenaunay-like syndrome

Prenatal onset

Detection of lymphatic abnormality in the prenatal period; isolated pedal edema excluded from this definition, because this may be a presentation of Milroy disease

Segment

A region of the body affected by lymphedema (for example, the face, conjunctiva, genitalia, upper limbs, lower limbs—each constitutes one body part); multisegmental refers to more than one segment affected by lymphedema (bilateral lower limb swelling not considered multisegmental lymphedema)

Syndromic

A constellation of abnormalities, one of which is lymphedema

Systemic involvement

Systemic lymphatic problems persisting beyond the newborn period or manifesting at any age thereafter, including hydrops fetalis, chylous ascites, intestinal lymphangiectasia, pleural and pericardial effusions, and pulmonary lymphangiectasia

Vascular anomalies

Includes congenital vascular abnormalities

Primary Lymphedema From Genes Involved in Lymphangiogenesis VEGFR-3 Mutations Abnormalities within the gene that encodes VEGFR-3 on chromosome 5q35 cause Milroy disease.27,28 Mutations in VEGFR-3 (also known as FLT4) are found in 70% of patients with congenital onset primary lymphedema.29 Inheritance is autosomal dominant with 85% penetrance.27,30 De novo mutations may occur, and thus a family history is not mandatory. Animal models have confirmed the role of VEGFR-3 in lymphangiogenesis, and studies have demonstrated that mutant VEGFR-3 showed impaired kinase activity.31,32 To date all reported VEGFR-3 mutations have occurred within the tyrosine kinase domain, but the exact mechanism of disease is not yet fully understood. An investigation of patients with lymphoscintigraphy suggests the failure of initial lymphatic vessel absorption and demonstrates a characteristic picture of “functional aplasia of lymphatic vessels.”25 Unlike the mouse model (Chy-mouse),33 in humans the initial lymphatic vessels are present (confirmed on histologic examination) but presumably unable to absorb interstitial fluid.34 It is clear how VEGFR-3 mutations may cause lymphatic dysplasia given its key role in the lymphangiogenesis pathway. However, further information on the exact mechanism of disease is necessary to optimize future treatments.

VEGF-C Mutations VEGF-C is a ligand for VEGFR-3 and controls lymphatic sprouting during embryonic development.35,36 Two families with congenital lymphedema of the lower limb had frameshift mutations in the VEGF-C gene on chromosome 4q34.37,38 It seems logical that mutations in the ligand for

K23349_Neligan_08_Genetics_r4_dc_0121-0134.indd 127

5/27/2015 9:20:20 AM

128

Part II  Anatomy, Physiology, and Lymphangiogenesis

VEGFR-3 will cause the same problems as a mutation in VEGFR-3 (for example, Milroy disease). In vivo overexpression assays in zebrafish confirm that VEGF-C frameshift mutations are causative for the Milroy-like phenotype seen.37 Therefore it is appropriate to consider VEGF-C screening in patients with Milroy-like lymphedema who have negative screening results for VEGFR-3 mutations.

CCBE1 Mutations Mutations in CCBE1 on chromosome 18q21 have been identified as causal for many patients with autosomal recessive, generalized lymphatic dysplasia (for example, Hennekam syndrome).39,40 Nearly all reported CCBE1 mutations have been missense, but one frameshift mutation has been reported.39,40 The phenotype composes lymphedema of all four limbs, lymphangiectasia (dysplasia of the lymphatic system of the intestines and/or lungs), variable degrees of learning difficulties, and characteristic facies (a flat face, flat and broad nasal bridge, and hypertelorism). Other associated problems may include hypothyroidism, glaucoma, seizures, hearing loss, and renal abnormalities. In severe cases it may present in the antenatal period with hydrops fetalis and/or cardiac abnormalities.41,42 Lymphoscintigraphy has rarely been undertaken in this condition. However, Bellini et al42 demonstrated abnormal drainage in the upper and lower limbs and the thoracic duct in one patient. Several affected individuals have a positive family history of lymphedema, which is usually suggestive of autosomal recessive inheritance inferring a higher risk of recurrence. CCBE1 mutations were present in only 23% of reported cases of Hennekam syndrome, which suggests genetic heterogeneity within this group.39 This is supported by the recent identification of homozygous or compound heterozygous mutations of the FAT4 gene (a member of the protocadherin family and not a recognized component of the lymphangiogenesis pathway) in 4 of 24 patients with CCBE1 mutation–negative Hennekam syndrome.43 CCBE1 plays a crucial role in lymphangiogenesis, including enhancing the effects of VEGF-C and subsequent receptor signaling. Therefore it is understandable that CCBE1 mutations cause widespread and severe abnormalities of lymphatic function.

SOX18 Mutations Three families with hypotrichosis-telangiectasia-lymphedema syndrome resulting from mutations in the SOX18 gene have been described.44 Two of the families were consanguineous with homozygous missense mutations in the SOX18 gene, which was located in 20q13. In the third family, the parents were nonconsanguineous, and both the affected child and his brother (who died in utero of hydrops fetalis) showed a heterozygous nonsense mutation that was not found in genomic DNA from either parent and constituted a de novo germline mutation. This rare form of primary lymphedema occurs as a result of either autosomal recessive or dominant SOX18 mutations negatively impacting on lymphatic development. The exact mechanism of disease remains unclear.

K23349_Neligan_08_Genetics_r4_dc_0121-0134.indd 128

5/27/2015 9:20:20 AM

Chapter 8  Impact of Genetics on Lymphangiogenesis

129

FOXC2 Mutations Lymphedema-distichiasis syndrome is inherited in an autosomal dominant manner and occurs as a result of mutations in the FOXC2 gene on chromosome 16q24.45 Brice et al46 demonstrated that mutations can be identified in more than 95% of patients with typical lymphedema-distichiasis syndrome, such as late-onset lower limb lymphedema and distichiasis (aberrant eyelashes arising from the meibomian glands). Many cases result from small deletions or insertions.47 FOXC2 encodes a transcription factor necessary for the signal transduction pathway, which ensures normal development of the lymphatic collecting vessels and valves.48 Given the role of FOXC2, it is obvious how mutations in the FOXC2 gene will cause clinical signs of impaired lymphatic function. Lymphoscintigraphic findings of affected individuals demonstrated reflux of lymph within the lower limbs as a result of valve failure within the hyperplastic lymphatics.22 Similarly, abnormal venous valves led to early-onset venous reflux in all patients with FOXC2 mutations.49

Primary Lymphedema Resulting From Mutations in Genes Indirectly With Unknown Involvement in Lymphangiogenesis Several subtypes of primary lymphedema occur as a result of abnormalities within genes not known to be part of the lymphangiogenesis pathway. Some of these conditions will be discussed here. In time it may become evident how these genes impact on lymphatic development.

GATA2 Mutations Mutations in the GATA2 gene on chromosome 3q21 cause Emberger syndrome and are inherited in an autosomal dominant manner but with a high rate of new mutations.50 GATA2 is expressed in lymphatic, vascular, and endocardial endothelial cells.51 Mouse studies suggest the programming of lymphatic valve development may be affected as a result of loss of function of Gata2.52 GATA2 is also involved in the regulation of hematopoiesis, and mutations have been reported in heritable forms of immunodeficiency, myelodysplastic syndrome, and acute myeloid leukemia.53,54 This explains the phenotypic association of late-onset primary lymphedema, severe cutaneous warts, and myelodysplastic syndrome and/or acute myeloid leukemia.55 However, the association with high-frequency, progressive sensorineural deafness is harder to explain.

AKT1 Pathway Mutations Lymphedema may be seen in conjunction with vascular abnormalities, disorders of growth, and cutaneous abnormalities, including Proteus syndrome. This progressive overgrowth disorder is characterized by progressive, segmental overgrowth of skin, connective tissue, fat, skeletal, and central nervous systems. Lymphatic and capillary malformations are commonly observed vascular changes. Manifestations of Proteus syndrome may not be present at birth but develop and progress during infancy. The diagnosis can be established with diagnostic clinical criteria and/or molecular analysis.56 Most individuals with clinically confirmed typical Proteus syndrome are identified to have a common activating mutation in AKT1, c.49G.A (p.Glu17Lys), arising as a result of somatic mosaicism. It is hypothesized that germline AKT1 c.49G.A mutations would be lethal early in embryonic development.57 The exact mechanism of disease is unclear, but mouse models suggest that Akt1 is required for lymphatic network formation, remodeling, and valve development.58

K23349_Neligan_08_Genetics_r4_dc_0121-0134.indd 129

5/27/2015 9:20:20 AM

130

Part II  Anatomy, Physiology, and Lymphangiogenesis

AKT acts within a molecular pathway that includes the mTOR and PIK3CA genes. This pathway is critical as a progrowth and antiapoptosis facilitator in cancer, but mutations in these genes have other implications. PIK3CA mutations have also been implicated in a range of disorders associated with somatic overgrowth, vascular malformations, fibroadipose overgrowth, pigmented cutaneous lesions, and megalencephaly.59-63 The identification of the PIK3/AKT/mTOR signaling pathway underpinning the disease processes gives rise to the potential for therapeutic targets through inhibition of this pathway.61

Syndromic Conditions Lymphedema is a recognized feature of many syndromes. In these conditions the lymphedema is not the primary problem but is an associated feature. The genetic causes of many of these syndromes are known and testing is available. A molecular result means that the mode of inheritance can be established, and where indicated, prenatal diagnosis can be offered. Turner and Noonan are the syndromes most frequently associated with lymphedema.25 Turner syndrome should always be considered as the cause of swelling in female infants presenting with congenital lymphedema of the hands and/or feet. Turner syndrome occurs as a result of a loss of one X chromosome. How this affects lymphangiogenesis remains unclear.

Microcephaly With or Without Chorioretinopathy, Lymphedema, and Intellectual Disability An autosomal dominant syndrome composing microcephaly, chorioretinopathy, and congenital lymphedema (mimicking Milroy disease both clinically and on investigation with lymphoscintigraphy) occurs as a result of mutations in the KIF11 gene on chromosome 10q24.64 The KIF11 protein product EG5 (a kinesin motor protein) is involved in spindle formation in mitosis but is not known to be directly involved in the lymphangiogenesis pathway. The mechanism by which KIF11/EG5 exerts its influence on the development of lymphatics remains a mystery.

Late-Onset Lower Limb or Four-Limb Lymphedema Mutations in the gap junction protein, gamma 2 (GJC2) gene have been implicated in the development of late-onset lymphedema of both lower limbs or all four limbs.65 The age of swelling onset varies considerably, ranging from childhood up to the sixth decade.66 GJC2 encodes the connexin-47 protein located on chromosome 1q42. The functional role of GJC2 within the lymphatic system is unclear. Lymphoscintigraphic findings performed in affected individuals showed lymphatic tracts that appear normal, but when quantification was done, there was significantly reduced absorption from tissues by the peripheral lymphatics in all four limbs.66 Therefore it can be hypothesized that normal GJC2 function is not required for lymphatic vessel development, but perhaps is necessary for maintenance of lymphatic function. This phenotype could explain the considerable number of patients presenting in adulthood with lymphedema without an obvious cause of their presumed “secondary lymphedema.” Clinicians should always consider a genetically determined primary lymphatic weakness in the absence of a clear underlying secondary cause. Imaging, such as with lymphoscintigraphy, may prove helpful in the investigation. For example, if the patient presents with unilateral lower limb swelling

K23349_Neligan_08_Genetics_r4_dc_0121-0134.indd 130

5/27/2015 9:20:21 AM

Chapter 8  Impact of Genetics on Lymphangiogenesis

131

but lymphoscintigraphy shows bilateral impaired lower limb lymphatic drainage, then the likely cause of the lymphedema will be an intrinsic primary lymphatic abnormality rather than an external secondary cause.

Conclusion Numerous causal genes have now been identified in the pathogenesis of primary lymphedema, most within the past few years. Gene identification helps delineate the phenotype more specifically. The identification of the genetic causes of primary lymphedema provides a molecular diagnostic test for some of the subtypes. Patients and families benefit hugely from a molecular diagnosis, because it allows the clinician to confidently predict the clinical prognosis and offer screening for family members. It is clear how some primary lymphedema subtypes are caused by abnormal gene function (for example, VEGFR-3 mutations and Milroy disease; CCBE1 and Hennekam syndrome), because the causal genes play a significant role in the lymphangiogenic pathway. However, genes implicated in other forms of primary lymphedema are not known to play a role in lymphangiogenesis. For example, how does a mutation in KIF11 cause microcephaly with or without chorioretinopathy, lymphedema, and intellectual disability? It is clear that we have more to learn about lymphangiogenesis and the genes that regulate it. The identification of more pathogenic genes will advance our knowledge of the etiopathogenesis of lymphatic disease. In time this may allow the development of improved therapeutic interventions. For example, gene therapy may become a feasible treatment strategy for patients with primary lymphedema.

R EFERENCES 1. Martinez-Corral I, Makinen T. Regulation of lymphatic vascular morphogenesis: implications for pathological (tumor) lymphangiogenesis. Exp Cell Res 319:1618-1625, 2013. 2. Oliver G, Alitalo K. The lymphatic vasculature: recent progress and paradigms. Annu Rev Cell Dev Biol 21:457-483, 2005. 3. Okuda KS, Astin JW, Misa JP, et al. Iyve1 expression reveals novel lymphatic vessels and new mechanisms for lymphatic vessel development in zebrafish. Development 139:2381-2391, 2012. 4. Oliver G. Lymphatic vasculature development. Nat Rev Immunol 4:35-45, 2004. 5. Schulte-Merker S, Sabine A, Petrova TV. Lymphatic vascular morphogenesis in development, physiology, and disease. J Cell Biol 193:607-618, 2011. 6. Wigle JT, Harvey N, Detmar M, et al. An essential role for Prox1 in the induction of the lymphatic endothelial cell phenotype. EMBO J 21:1505-1513, 2002. 7. Tammela T, Petrova TV, Alitalo K. Molecular lymphangiogenesis: new players. Trends Cell Biol 15:434441, 2005. 8. François M, Caprini A, Hosking B, et al. Sox18 induces development of the lymphatic vasculature in mice. Nature 456:643-647, 2008. 9. Lee S, Kang J, Yoo J, et al. Prox1 physically and functionally interacts with COUP-TFII to specify lymphatic endothelial cell fate. Blood 113:1856-1859, 2009.

K23349_Neligan_08_Genetics_r4_dc_0121-0134.indd 131

5/27/2015 9:20:21 AM

132

Part II  Anatomy, Physiology, and Lymphangiogenesis

10. You LR, Lin FJ, Lee CT, et al. Suppression of Notch signalling by the COUP-TFII transcription factor regulates vein identity. Nature 435:98-104, 2005. 11. Yang Y, García-Verdugo JM, Soriano-Navarro M, et al. Lymphatic endothelial progenitors bud from the cardinal vein and intersomitic vessels in mammalian embryos. Blood 120:2340-2348, 2012. 12. Hagerling R, Pollmann C, Andreas M, et al. A novel multistep mechanism for initial lymphangiogenesis in mouse embryos based on ultramicroscopy. EMBO J 32:629-644, 2013. 13. Veikkola T, Jussila L, Makinen T, et al. Signalling via vascular endothelial growth factor receptor-3 is sufficient for lymphangiogenesis in transgenic mice. EMBO J 20:1223-1231, 2001. 14. Karkkainen MJ, Haiko P, Sainio K, et al. Vascular endothelial growth factor C is required for sprouting of the first lymphatic vessels from embryonic veins. Nat Immunol 5:74-80, 2004. 15. Le Guen L, Karpanen T, Schulte D, et al. CCBE1 regulates VEGFC-mediated induction of VEGFR3 signaling during embryonic lymphangiogenesis. Development 141:1239-1249, 2014. 16. Jeltsch M, Jha SK, Tvorogov D, et al. CCBE1 enhances lymphangiogenesis via A disintegrin and metalloprotease with thrombospondin motifs-3-mediated vascular endothelial growth factor-C activation. Circulation 129:1962-1971, 2014. 17. Xu Y, Yuan L, Mak J, et al. Neuropilin-2 mediates VEGF-C-induced lymphatic sprouting together with VEGFR3. J Cell Biol 188:115-130, 2010. 18. Shawber CJ, Funahashi Y, Francisco E, et al. Notch alters VEGF responsiveness in human and murine endothelial cells by direct regulation of VEGFR-3 expression. J Clin Invest 117:3369-3382, 2007. 19. Baluk P, Fuxe J, Hashizume H, et al. Functionally specialized junctions between endothelial cells of lymphatic vessels. J Exp Med 204:2349-2362, 2007. 20. Sabine A, Agalarov Y, Maby-El Hajjami H, et al. Mechanotransduction, PROX1, and FOXC2 cooperate to control connexin37 and calcineurin during lymphatic-valve formation. Dev Cell 22:430-445, 2012. 21. Bazigou E, Makinen T. Flow control in our vessels: vascular valves make sure there is no way back. Cell Mol Life Sci 70:1055-1066, 2013. 22. Petrova TV, Karpanen T, Norrmén C, et al. Defective valves and abnormal mural cell recruitment underlie lymphatic vascular failure in lymphedema distichiasis. Nat Med 10:974-981, 2004. 23. Bridenbaugh EA, Gashev AA, Zawieja DC. Lymphatic muscle: a review of contractile function. Lymphat Res Biol 1:147-158, 2003. 24. Connell F, Kalidas K, Ostergaard P, et al. Linkage and sequence analysis indicate that CCBE1 is mutated in recessively inherited generalised lymphatic dysplasia. Hum Genet 127:231-241, 2010. 25. Connell FC, Gordon K, Brice G, et al. The classification and diagnostic algorithm for primary lymphatic dysplasia: an update from 2010 to include molecular findings. Clin Genet 84:303-314, 2013. 26. Mendola A, Schlögel MJ, Ghalamkarpour A, et al. Mutations in the VEGFR3 signaling pathway explain 36% of familial lymphedema. Mol Syndromol 4:257-266, 2013. 27. Ferrell RE, Levinson KL, Esman JH, et al. Hereditary lymphedema: evidence for linkage and genetic heterogeneity. Hum Mol Genet 7:2073-2078, 1998. 28. Gordon K, Spiden SL, Connell FC, et al. FLT4/VEGFR3 and Milroy disease: novel mutations, a review of published variants and database update. Hum Mutat 34:23-31, 2013. 29. Connell FC, Ostergaard P, Carver C, et al. Analysis of the coding regions of VEGFR3 and VEGFC in Milroy disease and other primary lymphoedemas. Hum Genet 124:625-631, 2009. 30. Evans AL, Brice G, Sotirova V, et al. Mapping of primary congenital lymphedema to the 5q35.3 region. Am J Hum Genet 64:547-555, 1999. 31. Kaipainen A, Korhonen J, Mustonen T, et al. Expression of the FMS-like tyrosine kinase 4 gene becomes restricted to lymphatic endothelium during development. Proc Natl Acad Sci U S A 92:35663570, 1995. 32. Karkkainen MJ, Ferrell RE, Lawrence EC, et al. Missense mutations interfere with VEGFR-3 signalling in primary lymphoedema. Nat Genet 25:153-159, 2000.

K23349_Neligan_08_Genetics_r4_dc_0121-0134.indd 132

5/27/2015 9:20:21 AM

Chapter 8  Impact of Genetics on Lymphangiogenesis

133

33. Karkkainen MJ, Saaristo A, Jussila L, et al. A model for gene therapy of human hereditary lymphedema. Proc Natl Acad Sci U S A 98:12677-12682, 2001. 34. Mellor RH, Hubert CE, Stanton AW, et al. Lymphatic dysfunction, not aplasia, underlies Milroy disease. Microcirculation 17:281-296, 2010. 35. Küchler AM, Gjini E, Peterson-Maduro J, et al. Development of the zebrafish lymphatic system requires VEGFC signaling. Curr Biol 16:1244-1248, 2006. 36. Hogan BM, Bos FL, Bussmann J, et al. CCBE1 is required for embryonic lymphangiogenesis and venous sprouting. Nat Genet 41:396-398, 2009. 37. Gordon K, Schulte D, Brice G, et al. Mutation in vascular endothelial growth factor-C, a ligand for vascular endothelial growth factor receptor-3, is associated with autosomal dominant milroy-like primary lymphedema. Circ Res 112:956-960, 2013. 38. Balboa-Beltran E, Fernández-Seara MJ, Pérez-Muñuzuri A, et al. A novel stop mutation in the vascular endothelial growth factor-C gene (VEGFC) results in Milroy-like disease. J Med Genet 51:475-478, 2014. 39. Alders M, Hogan BM, Gjini E, et al. Mutations in CCBE1 cause generalized lymph vessel dysplasia in humans. Nat Genet 41:1272-1274, 2009. 40. Connell FC, Kalidas K, Ostergaard P, et al. CCBE1 mutations can cause a mild, atypical form of generalized lymphatic dysplasia but are not a common cause of non-immune hydrops fetalis. Clin Genet 81:191-197, 2012. 41. Van Balkom ID, Alders M, Allanson J, et al. Lymphedema-lymphangiectasia-mental retardation (Hennekam) syndrome: a review. Am J Med Genet 112:412-421, 2002. 42. Bellini C, Mazzella M, Arioni C, et al. Hennekam syndrome presenting as nonimmune hydrops fetalis, congenital chylothorax, and congenital pulmonary lymphangiectasia. Am J Med Genet A 120A:92-96, 2003. 43. Alders M, Al-Gazali L, Cordeiro I, et al. Hennekam syndrome can be caused by FAT4 mutations and be allelic to Van Maldergem syndrome. Hum Genet 133:1161-1167, 2014. 44. Irrthum A, Devriendt K, Chitayat D, et al. Mutations in the transcription factor gene SOX18 underlie recessive and dominant forms of hypotrichosis-lymphedema-telangiectasia. Am J Hum Genet 72:14701478, 2003. 45. Fang J, Dagenais SL, Erickson RP, et al. Mutations in FOXC2 (MFH-1), a forkhead family transcription factor, are responsible for the hereditary lymphedema-distichiasis syndrome. Am J Hum Genet 67:1382-1388, 2000. 46. Brice G, Mansour S, Bell R, et al. Analysis of the phenotypic abnormalities in lymphoedema-distichiasis syndrome in 74 patients with FOXC2 mutations or linkage to 16q24. J Med Genet 39:478-483, 2002. 47. Bell R, Brice G, Child AH, et al. Analysis of lymphoedema-distichiasis families for FOXC2 mutations reveals small insertions and deletions throughout the gene. Hum Genet 108:546-551, 2001. 48. Norrmén C, Ivanov KI, Cheng J, et al. FOXC2 controls formation and maturation of lymphatic collecting vessels through cooperation with NFATc1. J Cell Biol 185:439-457, 2009. 49. Mellor RH, Brice G, Stanton AW, et al. Mutations in FOXC2 are strongly associated with primary valve failure in veins of the lower limb. Circulation 115:1912-1920, 2007. 50. Ostergaard P, Simpson MA, Brice G, et al. Rapid identification of mutations in GJC2 in primary lymphoedema using whole exome sequencing combined with linkage analysis with delineation of the phenotype. J Med Genet 48:251-255, 2011. 51. Khandekar M, Brandt W, Zhou Y, et al. A GATA2 intronic enhancer confers its pan-endothelia-specific regulation. Development 134:1703-1712, 2007. 52. Kazenwadel J, Secker GA, Liu YJ, et al. Loss-of-function germline GATA2 mutations in patients with MDS/AML or MonoMAC syndrome and primary lymphedema reveal a key role for GATA2 in the lymphatic vasculature. Blood 119:1283-1291, 2012.

K23349_Neligan_08_Genetics_r4_dc_0121-0134.indd 133

5/27/2015 9:20:21 AM

134

Part II  Anatomy, Physiology, and Lymphangiogenesis

53. Hahn CN, Chong CE, Carmichael CL, et al. Heritable GATA2 mutations associated with familial myelodysplastic syndrome and acute myeloid leukemia. Nat Genet 43:1012-1017, 2011. 54. Hsu AP, Sampaio EP, Khan J, et al. Mutations in GATA2 are associated with the autosomal dominant and sporadic monocytopenia and mycobacterial infection (MonoMAC) syndrome. Blood 118:26532655, 2011. 55. Mansour S, Connell F, Steward C, et al. Emberger syndrome—primary lymphedema with myelodysplasia: report of seven new cases. Am J Med Genet A 152A:2287-2296, 2010. 56. Biesecker L. The challenges of Proteus syndrome: diagnosis and management. Eur J Hum Genet 14:151157, 2006. 57. Lindhurst MJ, Sapp JC, Teer JK, et al. A mosaic activating mutation in AKT1 associated with the Proteus syndrome. N Engl J Med 365:611-619, 2011. 58. Zhou F, Chang Z, Zhang L, et al. Akt/Protein kinase B is required for lymphatic network formation, remodeling, and valve development. Am J Pathol 177:2124-2133, 2010. 59. Kurek KC, Luks VL, Ayturk UM, et al. Somatic mosaic activating mutations in PIK3CA cause CLOVES syndrome. Am J Hum Genet 90:1108-1115, 2012. 60. Lee JH, Huynh M, Silhavy JL, et al. De novo somatic mutations in components of the PI3K-AKT3mTOR pathway cause hemimegalencephaly. Nat Genet 44:941-945, 2012. 61. Lindhurst MJ, Parker VE, Payne F, et al. Mosaic overgrowth with fibroadipose hyperplasia is caused by somatic activating mutations in PIK3CA. Nat Genet 44:928-933, 2012. 62. Rivière JB, Mirzaa GM, O’Roak BJ, et al. De novo germline and postzygotic mutations in AKT3, PIK3R2 and PIK3CA cause a spectrum of related megalencephaly syndromes. Nat Genet 44:934-940, 2012. 63. Emrick LT, Murphy L, Shamshirsaz AA, et al. Prenatal diagnosis of CLOVES syndrome confirmed by detection of a mosaic PIK3CA mutation in cultured amniocytes. Am J Med Genet A 164A:2633-2637, 2014. 64. Ostergaard P, Simpson MA, Mendola A, et al. Mutations in KIF11 cause autosomal-dominant microcephaly variably associated with congenital lymphedema and chorioretinopathy. Am J Hum Genet 90:356-362, 2012. 65. Ferrell RE, Baty CJ, Kimak MA, et al. GJC2 missense mutations cause human lymphedema. Am J Hum Genet 86:943-948, 2010. 66. Ostergaard P, Simpson MA, Brice G, et al. Rapid identification of mutations in GJC2 in primary lympho­edema using whole exome sequencing combined with linkage analysis with delineation of the phenotype. J Med Genet 48:251-255, 2011.

K23349_Neligan_08_Genetics_r4_dc_0121-0134.indd 134

5/27/2015 9:20:21 AM

C hapter 9 Relationship Between Fat Tissue and Lymphangiogenesis Mauro Andrade

K ey P oints • Adipose tissue overgrowth is a key clinical feature in chronic lymphedema. • Lymphangiogenesis may play a critical role in lymphedema-related adipogenesis. • It is still unclear how the intricate mechanisms of lymphangiogenesis, inflammation, and adipogenesis are interrelated.

A Fat deposition has long been recognized as one of the clinical features of elephantiasis. In a very elegant clinical description, Moritz Kaposi,1 in the nineteenth century, stated: “If an incision is made on a limb with advanced elephantiasis . . . the subcutaneous tissue is increased many fold . . .” In the twentieth century, debulking procedures to remove excess skin and fat became popular as the preferred surgical treatment for patients with lymphedema.

Rel

Until recently, much of our knowledge about the pathophysiology of lymph stasis focused on protein retention, altered immune cell trafficking, and inflammation. Little attention was directed to fat deposition, notwithstanding the fact that some authors considered it a subproduct of the inflammatory response.2 In the late 1990s, a great discussion ensued after Brorson and Svensson’s publication of a series of patients treated with liposuction.3 Soon after, advances in the comprehension of molecular mechanisms related to lymphangiogenesis and adipogenesis provided new insights into the relationship between lymph stasis and fat. This chapter discusses how these findings have influenced our current knowledge of the relationship between fat tissue and lymphangiogenesis and how this understanding can lead to future therapeutic approaches in patients with lymphedema. 135

K23346_Neligan_09_Relationship_r2_dc_0135-0142.indd 135

5/26/2015 8:52:50 AM

136

Part II  Anatomy, Physiology, and Lymphangiogenesis

Adipogenesis For a long time, the known functions of subcutaneous fat were limited to energy storage and regulation of body temperature. Perhaps because of its relatively simple structure, adipose tissue was a victim of several decades of scientific neglect. It was not until the more recent recognition of the burden of obesity in modern society that further investigation into the nature of adipose tissue unveiled a whole complex and dynamic physiologic organ. Adipogenesis is the process of differentiation that turns preadipocytes or multipotent stem cells into mature adipocytes. This is regulated by several mechanisms (transcription factors, cofactors, and signaling) from as early as the embryonic period until adult life. The number of adipocytes does not vary after childhood and adolescence, even after weight changes.4 Thus most weight gain or loss depends on the size variation of the intracellular storage of triglycerides and cholesteryl esters in fat droplets. Because the total population of fat cells remains constant in adulthood, new adipocytes can be generated by two different pathways: preexisting undifferentiated progenitor cells or through dedifferentiation of mature adipocytes into preadipocytes, which can proliferate and differentiate into mature adipocytes.5 The annual turnover rate of fat cells is estimated to be 10%.4 The differentiation from a stem cell precursor to a mature adipocyte involves a complex sequence of gene expression and signaling and is subject to the environment in which it occurs, either in culture or in vivo. Multipotent stem cells are capable of differentiating into mesodermal cell types (adipocytes, chondrocytes, osteoblasts, and myocytes). Under stimulation by hormones, cytokines, and growth factors (insulin, insulin-like growth factor 1, glucocorticoids, mineralocorticoids, and thyroid hormone), some multipotent stem cells become committed to the adipogenic lineage (preadipocytes). Preadipocytes then differentiate into adipocytes by the action of transcription factors that activate specific genes responsible for the adipocyte phenotype. The most important transcription factors at this phase are CCAAT/enhancer-binding protein alpha (C/EBP-alpha) and peroxisome proliferator-activated receptor gamma (PPAR-gamma).6 Fig. 9-1 shows the normal relationship between the lymphatic system and fat in a limb.

Lymphangiogenesis Although lymphangiogenesis is discussed in greater detail elsewhere in Chapter 7, some brief aspects of lymphatic system development will illustrate the relationship between the lymphatics and fat. The lymphatic system appears during the sixth and seventh weeks of embryonic development,7 4 weeks after the primary components of the blood circulation arise. Two different theories about the origin of the lymphatics were proposed at the beginning of the twentieth century. Sabin’s anatomic studies8 suggested that sprouting from blood endothelial cells was the origin of the lymphatic system. This was in opposition to Huntington’s theory of the centripetal formation of the lymphatics from mesenchymal lymphangioblasts, which reached the venous system later in development.9 Some controversies still remain, and evidence seems to support both theories.10,11

K23346_Neligan_09_Relationship_r2_dc_0135-0142.indd 136

5/26/2015 8:52:50 AM

Chapter 9  Relationship Between Fat Tissue and Lymphangiogenesis

Healthy

Acute lymphedema Generalized protein and fluid accumulation Inflammation Some dilation and fibrosis of lymphatics

137

Chronic lymphedema Sustained inflammatory response Increased and excessive adipose tissue Fibrosis and tissue architecture disruption Dilated and fibrosed lymphatics Lymphangiogenesis

FIG. 9-1  Left, Normal lymphatic circulation is demonstrated in a normal limb. Note the relationship between the lymphatics and the normal amount of fat in the limb. Center, A lymphedematous limb in acute lymphedema. Note the increased fat deposition compared with the healthy limb. Right, In a chronically lymphedematous limb there is increased fat deposition and increased fibrosis not only in the lymphatics, but also in the soft tissue of the limb.

The regulation and development of the lymphatic system depend on several signaling factors and cell receptors. The most important signaling factors are glycoproteins known as vascular endothelial growth factors (VEGFs). VEGFs are the primary regulators of endothelial proliferation, angiogenesis, vasculogenesis, and vascular permeability.12 There are six VEGF subtypes (A, B, C, D, and E) and placental growth factor (PlGF) that bind to specific membrane tyrosine kinase receptors. Three different VEGF tyrosine kinase receptors have been identified so far: • Vascular endothelial growth factor receptor 1 (VEGFR-1) (Flt-1) • VEGFR-2 (Flk-1 and KDR) • VEGFR-3 (Flt-4) VEGF-B and PlGF bind to VEGFR-1, whereas VEGF-A interacts with both VEGFR-1 and VEGFR-2. VEGF-E binds to VEGFR-2, and both VEGF-C and VEGF-D bind to VEGFR-3. VEGFR-1 and VEGFR-2 mediate angiogenesis, whereas VEGFR-3 is involved mainly in lymphangiogenesis. VEGFs and VEGFRs are essential for blood vessel development and angiogenesis. Unlike the other VEGFs, both VEGF-C and VEGF-D promote lymphangiogenesis through their specific lymphatic endothelial receptor VEGFR-3. Nevertheless, early experimental inactivation of Flt4, the gene responsible for encoding VEGFR-3, also results in defective blood vasculogenesis and

K23346_Neligan_09_Relationship_r2_dc_0135-0142.indd 137

5/26/2015 8:52:51 AM

138

Part II  Anatomy, Physiology, and Lymphangiogenesis

angiogenesis,13 demonstrating that VEGF-C not only has a critical role in the development of the lymphatic system but is also essential in the development of the embryonic cardiovascular system. Lymphangiogenesis is a developmental process that occurs mainly in the embryonic phase and is uncommon in adult tissue. In adults new lymphatic tissue is formed in wound healing and is induced by some tumors. Thus the exact role of VEGF-C and VEGF-D in normal human physiology is unclear. Some VEGF actions have been described: stimulation of growth of lymphatic capillaries and recruitment of inflammatory cells,14 regulation of endothelial fatty acid, and control of salt-dependent interstitial volume and blood pressure.15 Interleukin-7 (IL-7) and interleukin-1 beta are cytokines involved in lymphoid tissue regulation and inflammation and can also participate in lymphangiogenesis. IL-7 increases the expression of lymphatic markers (lymphatic vessel endothelial hyaluronan receptor 1 [LYVE-1], podoplanin, and Prox1) in endothelial cells, and it induces the formation of lymphatic vessels in vivo. Interleukin-1 beta upregulates VEGF-C, and IL-7 upregulates VEGFR-3 and the expression of VEGF-D in endothelial cells.16

Where the Pathways of Lymphangiogenesis and Adipogenesis Cross The concept that lymphedema is restricted to tissue fluid accumulation is long gone. Advances in research on molecular mechanisms involved in cancer metastasis and obesity revealed intimate pathways of lymphangiogenesis, inflammation, and adipogenesis and opened a broad new field in the investigation of lymphatic disorders. Also, a lack of available experimental models for lymphedema has hindered specific evaluation of how lymph stasis influences tissue changes. Recently an experimental model of lymphedema17,18 provided new insights into the mechanisms responsible for many of the events resulting from impaired clearance of tissue lymph. The blockage of the lymphatic pathways in either clinical settings or experimental models affects lymph cell trafficking that leads to inflammation, fibrosis, and the disruption of normal tissue architecture. It is the result of the continuous production of growth factors, proteolytic enzymes, angiogenic factors, peroxisome proliferator-activated receptors, acute phase proteins (serum amyloid P), components of the renin-angiotensin-aldosterone system (Ang II), and cytokines, similar to other fibrotic disorders.19 In chronic lymphedema, the subcutaneous tissue displays a constant histologic pattern of inflammatory cell accumulation, increased amounts of ground substance, fibrosis, dilated lymphatic vessels, and proliferation of adipose tissue, whereas acute lymph stasis is accompanied by a marked increase in monocytes and macrophages.20 In fact, unlike chronic lymphedema, experimental acute lymph stasis resembles an inflammatory condition20 without the corresponding tissue damage (fibrosis and excessive fat) that is observed later in the development of the clinical disorder. A simple explanation may be that inflammation is the key trigger in the development of all histopathologic changes caused by chronic lymph stasis; fibrosis and fat deposition are a consequence of sustained inflammatory reaction in the tissue.

K23346_Neligan_09_Relationship_r2_dc_0135-0142.indd 138

5/26/2015 8:52:51 AM

Chapter 9  Relationship Between Fat Tissue and Lymphangiogenesis

139

It is well established that the lymphatics regulate the inflammatory response by transporting leukocytes and antigen-presenting cells from the site of inflammation to the lymph nodes. In lymph stasis, such regulation is impaired, suggesting that in this situation, the inflammatory reaction depends mostly on the local activity of monocytes and macrophages. Also, lymphangiogenesis is increased at the sites of tissue inflammation and wound repair, and VEGF-C and VEGF-D are overexpressed in macrophages.21 In addition, transforming growth factor beta, a cytokine that modulates local inflammation, acts as an inhibitor of lymphangiogenesis in inflammatory settings. Its inhibition decreases fibrosis and stimulates lymphangiogenesis, even without increases in VEGF-C expression.18,22 These findings support the hypothesis that there is a strong relationship and simultaneity between inflammation and lymphangiogenesis. On the other hand, experimental lymphedema in transgenic mice lacking VEGFR-3 does not induce fibrosis or an increase in lipid content and inflammation.23 This experimental model suggests that lymphangiogenesis and its regulators are essential to promote initial inflammation and late tissue changes, such as fibrosis and adipogenesis. However, is inflammation the first step in lymphangiogenesis and fat deposition? Are they independent features that converge in late-stage lymphedema? Is it lymphangiogenesis and lymph stasis that make lymphedema so different from other fibrotic diseases regarding fat deposition? Recent articles try to answer these intriguing questions. In patients undergoing axillary lymph node clearance, obesity is an established major risk factor for the development of lymphedema, but the underlying mechanism is unknown. Although the relationship between obesity and inflammatory response has elicited many publications, the correlation between lymphangiogenesis and obesity is still poorly understood and controversial. Increased circulating VEGF-C levels have been reported in obese individuals, and it has been suggested that such findings could explain the increased risk of metastasis that has been described in some cancers.24 On the other hand, two recent articles observed the opposite results. One study showed that adipose tissue played a protective role against prostate cancer dissemination.25 Conversely, another experiment that had mice consuming a high-fat diet demonstrated increased metastasis of the lymph nodes after enhanced lymphangiogenesis mediated by macrophage activation.26 Our group27 reported that fat cells in culture displayed marked expression of VEGFR-3 on the cell surface. Although fat cells must dedifferentiate to grow in culture media, the markers belong to undifferentiated mesenchymal cells. VEGF-C and VEGF-D may play a role in adipose cell replication as a response to lymph stasis. In fact, increased levels of circulating VEGF-D were found in the peripheral blood in patients with lymphedema. This suggests that a systemic response may try to increase lymphangiogenesis in the clinical setting of lymphatic stasis28 that could also influence adipogenesis and inflammation. In addition, lymph induced a more complete differentiation in preadipocytes in vitro.29 From these data it can be postulated that there is also a local action of lymph blockage, which favors a more important recruitment of mesenchymal cells into the adipose lineage. Experimental models of lymphedema are subjected to some criticism, because they hardly resemble the clinical evolution of lymphatic blockage where time is concerned; nevertheless, they seem to be useful, because they mimic the histopathologic picture that is observed in chronic human lymphedema. In a recently described model with mice tails,18 lymphatic blockage was obtained

K23346_Neligan_09_Relationship_r2_dc_0135-0142.indd 139

5/26/2015 8:52:51 AM

140

Part II  Anatomy, Physiology, and Lymphangiogenesis

by resection of full-thickness skin, excising superficial lymphatics, and performing direct ligation of deep lymphatics. In this model, Zampell et al30 observed a significant change in the subcutaneous tissue caused by fat cell hypertrophy with the incremental size of fat droplets and also an increased number of adipocytes. However, there was a concomitant mononuclear cell inflammatory response, which made it difficult to ascertain whether fat deposition was a cause or a consequence of local inflammation. The same group31 demonstrated that CCAAT/C/EBP-alpha, PPAR-gamma, and adiponectin were highly expressed in the same experimental model of lymphedema. Again, C/EBP-alpha and PPAR-gamma are crucial for preadipocytes to turn into mature adipocytes.6 Adiponectin is a plasma protein secreted by adipose tissue,32,33 is downregulated in obesity states, and may influence lymphangiogenesis. In another recent study with the mouse tail model, adiponectin knockout mice had increased tail lymphedema and a lower number of lymphatics compared with wild-type mice.34 These findings could be reversed with the administration of adenoviral vectors encoding adiponectin. Furthermore, the same study demonstrated that adiponectin promoted the growth of human lymphatic endothelial cells in culture by the phosphorylation of AMP-activated protein kinase and endothelial nitric oxide synthesis.34 A possible mechanism of adiponectin control of lymphangiogenesis in vivo is the induction of VEGF-C expression in macrophages.35 However, in clinical lymphedema both adipogenesis and lymphangiogenesis coexist, and thus further studies are needed to verify whether lymphangiogenesis promoted by lymph stasis (or inflammation) interferes with adiponectin release by adipocytes.

Conclusion Recent advances in our knowledge of the intimate molecular mechanisms that may influence the development of chronic lymphedema and its accompanying tissue damage are the consequence of two major developments in medical research during the past years: (1) detailed information about the intimacy of lymphangiogenesis regulation obtained through studies of cancer biology and (2) the increasing interest in another important public health issue—obesity. It is still unclear how all the markers, signaling factors, and receptors that have been identified in tissues under the influence of lymph stasis orchestrate the evolution of the intricate events that follow the disruption of the lymphatic pathways. Another problem is to define how inflammation relates to and promotes lymphangiogenesis and adipose growth. Is inflammation the trigger, or are they intertwined players in the development of late tissue changes? A third and no less compelling issue is to identify the temporal sequence of events (if there is any) that may point to a targeted intervention to stop disease progression. However, most of our current and very recent knowledge is based on an experimental model that may not entirely translate into the clinical challenges faced by lymphedema patients and practitioners. Therefore, although this exciting subject still needs further investigation, its first steps promise to challenge how we currently address lymphedema and change our future approach to lymph stasis disorders.

K23346_Neligan_09_Relationship_r2_dc_0135-0142.indd 140

5/26/2015 8:52:51 AM

Chapter 9  Relationship Between Fat Tissue and Lymphangiogenesis

141

C linical P earls • Imaging and clinical evaluation of the fat content in limbs with lymphedema are essential to define the prognosis. • It is expected that control of adipogenesis and/or inflammation resulting from lymph stasis must be proved, as essential steps in lymphedema management.

R EFERENCES 1. Kaposi M. Leçons sur les Maladies de la Peau, vol 2. Paris: G Masson, 1881. 2. Földi E, Földi M. Lymphostatic diseases. In Földi M, Földi E, Kubik S, eds. Textbook of Lymphology for Physicians and Lymphedema Therapists. Munich: Urban & Fischer, 2003. 3. Brorson H, Svensson H. Complete reduction of lymphoedema of the arm by liposuction after breast cancer. Scand J Plast Reconstr Hand Surg 31:137-143, 1997. 4. Spalding KL, Arner E, Westermark PO, et al. Dynamics of fat cell turnover in humans. Nature 453:783787, 2008. 5. Moreno-Navarrete JM, Fernández-Real JM. Adipocyte differentiation. In Symonds ME, ed. Adipose Tissue Biology. New York: Springer-Verlag, 2012. 6. Mandrup S, Lane MD. Regulating adipogenesis. J Biol Chem 272:5367-5370, 1997. 7. Tille JC, Pepper MS. Hereditary vascular anomalies: new insights into their pathogenesis. Arterioscler Thromb Vasc Biol 24:1578-1590, 2004. 8. Sabin F. On the origin of the lymphatic system from the veins and the development of the lymph hearts and thoracic duct in the pig. Am J Anat 1:367-391, 1902. 9. Huntington GS, McClure CFW. The anatomy and development of the jugular sacs in the domestic cat (Felis domestica). Am J Anat 10:177-311, 1910. 10. Wigle JT, Oliver G. Prox1 function is required for the development of the murine lymphatic system. Cell 98:769-778, 1999. 11. Schneider M, Othman-Hassan K, Christ B, et al. Lymphangioblasts in the avian wing bud. Dev Dyn 216:311-319, 1999. 12. Ferrara N, Alitalo K. Clinical applications of angiogenic growth factors and their inhibitors. Nat Med 5:1359-1364, 1999. 13. Dumont DJ, Jussila L, Taipale J, et al. Cardiovascular failure in mouse embryos deficient in VEGF receptor-3. Science 282:946-949, 1998. 14. Wirzenius M, Tammela T, Uutela M, et al. Distinct vascular endothelial growth factor signals for lymphatic vessel enlargement and sprouting. J Exp Med 204:1431-1440, 2007. 15. Hagberg CE, Falkevall A, Wang X, et al. Vascular endothelial growth factor B controls endothelial fatty acid uptake. Nature 464:917-921, 2010. 16. Al-Rawi MAA, Mansel RE, Jiang WG. Lymphangiogenesis and its role in cancer. Histol Histopathol 20:283-298, 2005. 17. Tabibiazar R, Cheung L, Han J, et al. Inflammatory manifestations of experimental lymphatic insufficiency. PLoS Med 3:e254, 2006. 18. Clavin NW, Avraham T, Fernandez J, et al. TGF-beta1 is a negative regulator of lymphatic regeneration during wound repair. Am J Physiol Heart Circ Physiol 295:H2113-H2127, 2008. 19. Wynn TA. Cellular and molecular mechanisms of fibrosis. J Pathol 214:199-210, 2008.

K23346_Neligan_09_Relationship_r2_dc_0135-0142.indd 141

5/26/2015 8:52:51 AM

142

Part II  Anatomy, Physiology, and Lymphangiogenesis

20. Piller NB. Macrophage and tissue changes in the developmental phases of secondary lymphoedema and during conservative therapy with benzopyrone. Arch Histol Cytol 53 Suppl:209-218, 1990. 21. Cursiefen C, Chen L, Borges LP, et al. VEGF-A stimulates lymphangiogenesis and hemangiogenesis in inflammatory neovascularization via macrophage recruitment. J Clin Invest 113:1040-1050, 2004. 22. Oka M, Iwata C, Suzuki HI, et al. Inhibition of endogenous TGF-beta signaling enhances lymphangiogenesis. Blood 111:4571-4579, 2008. 23. Markhus CE, Karlsen TV, Wagner M, et al. Increased interstitial protein because of impaired lymph drainage does not induce fibrosis and inflammation in lymphedema. Arterioscler Thromb Vasc Biol 33:266-274, 2013. 24. Silha JV, Krsek M, Sucharda P, et al. Angiogenic factors are elevated in overweight and obese individuals. Int J Obes (Lond) 29:1308-1314, 2005. 25. Moreira A, Pereira SS, Machado CL, et al. Obesity inhibits lymphangiogenesis in prostate tumors. Int J Clin Exp Pathol 7:348-352, 2013. 26. Jung JI, Cho HJ, Jung YJ, et al. High-fat diet-induced obesity increases lymphangiogenesis and lymph node metastasis in the B16F10 melanoma allograft model: roles of adipocytes and M2-macrophages. Int J Cancer 136:258-270, 2015. 27. Andrade M, Akamatsu F, Jacomo A. Adipocytes express specific lymphangiogenesis membrane receptors. FASEB J 25(Suppl):684.1, 2011. 28. Fink AM, Kaltenegger I, Schneider B, et al. Serum level of VEGF-D in patients with primary lymphedema. Lymphology 37:185-189, 2004. 29. Nougues J, Reyne Y, Dulor JP. Differentiation of rabbit adipocytes precursors in primary culture. Int J Obes 12:321-333, 1998. 30. Zampell JC, Aschen S, Weitman ES, et al. Regulation of adipogenesis by lymphatic fluid stasis: part I. Adipogenesis, fibrosis, and inflammation. Plast Reconstr Surg 129:825-834, 2012. 31. Aschen S, Zampell JC, Elhadad S, et al. Regulation of adipogenesis by lymphatic fluid stasis: part II. Expression of adipose differentiation genes. Plast Reconstr Surg 129:838-847, 2012. 32. Makki K, Froguel P, Wolowczuk I. Adipose tissue in obesity-related inflammation and insulin resistance: cells, cytokines, and chemokines. ISRN Inflamm 2013:139239, 2013. 33. Piya MK, McTernan PG, Kumar S. Adipokine inflammation and insulin resistance: the role of glucose, lipids and endotoxin. J Endocrinol 216:T1-T15, 2013. 34. Shimizu Y, Shibata R, Ishii M, et al. Adiponectin-mediated modulation of lymphatic vessel formation and lymphedema. J Am Heart Assoc 2:e000438, 2013. 35. Hu D, Fukuhara A, Miyata Y, et al. Adiponectin regulates vascular endothelial growth factor-C expression in macrophages via Syk-ERK pathway. PLoS One 8:e56071, 2013.

K23346_Neligan_09_Relationship_r2_dc_0135-0142.indd 142

5/26/2015 8:52:51 AM

C hapter 10 Lymphatic Malformations Sandro Michelini, Alessandro Fiorentino, Marco Cardone

K ey P oints • Vascular malformations are mistakes in the development of the vascular system; hemangiomas are vascular tumors. • Endothelial hyperplasia is not a characteristic of vascular malformations. • Vascular malformations have multiple causes and complex clinical presentations; most vascular malformations are mixed. • Lymphoscintigraphy represents the highest standard in diagnostic tests for primary and secondary lymphedema (see Chapter 26).

A

• Arteriography and venography are needed for patients who require surgical or endovascular treatment. • Surgical treatment is reserved for more complex forms of vascular malformations, especially those with low flow.

Lym Lymphatic malformations are a type of morphostructural vascular disease. They have been described and classified in several ways because of their different clinical manifestations and types of onset. Simple malformations are usually named in Latin (for example, hemangioma simplex, angioma telangiectaticum, and hemangioma cavernosum), whereas complex or mixed malformations are named for the authors who first described them (for example, Klippel-Trenaunay syndrome [leg length discrepancy, varicose veins, or erythema without evidence of arteriovenous fistulas] and Parkes-Weber syndrome [Klippel-Trenaunay syndrome with the presence of arteriovenous fistulas]). The incidence of vascular malformations is 1.5% worldwide1; the prevalence of venous forms is 1 child in 5000 or 10,000, depending on the study.2 In his 1876 treatise on the pathology of tumors, Virchow3 discussed the first accurate medical description of a vascular malformation involving a cirsoid aneurysm, dating back to the sixteenth 143

K23346_Neligan_10_Lymph Malform_r2_0143-0162.indd 143

5/26/2015 8:54:58 AM

144

Part II  Anatomy, Physiology, and Lymphangiogenesis

century. Nicoladoni4 and Branham5 nearly simultaneously described the occurrence of bradycardia after compression of a high-flow arteriovenous fistula. In 1964 Malan and Puglionisi6 published the first useful classification of angiodysplasia. Szilagyi et al7 further described clinical diagnosis and a therapeutic approach in 1976. In articles published in 1982, Mulliken and colleagues8,9 proposed a classification that distinguished between malformations or vascular abnormalities and vascular tumors (hemangiomas). This is currently the most widely used system. Vascular malformations are mistakes in the development of the vascular system; hemangiomas are vascular tumors. Mulliken et al9 described the characteristics of the endothelium and the biology of vascular malformations and hemangiomas. Approximately 30% of hemangiomas are present at birth, and 70% occur in the first 3 months of life. They grow during the proliferative phase (usually the first year of life) and regress in subsequent years. The female/male ratio of hemangiomas is 5:1. In a biologic study, hemangiomas had endothelial hyperplasia and an increase in mast cell inclusions; the cells grew if cultured, and they incorporated 3H-thymidine in their proliferative phase.10 Most hemangiomas regress spontaneously (95% are reduced by about 7 years of age) and require no therapy because any intervention (removal or reduction) would be aesthetically damaging due to healing of the tissues. In contrast, endothelial hyperplasia is not a characteristic of vascular malformations. These lesions do not have a proliferative phase (that is, they do not grow if placed in culture) and do not incorporate 3H-thymidine. They showed no mast cell inclusions in the biologic study.10 Their endothelium is surrounded by hyperplasia of the vascular wall. Approximately 90% of cases are present at birth, and the female/male ratio is 1:1. The growth of vascular malformations is related to the child’s growth and hemodynamic factors.

Embryology Vascular System The first channels of the vascular system can be seen beginning in the third week of gestation (see Chapter 4, Embryology). In 1922 Woollard11 sketched three stages of growth and differentiation: • Stage 1, the undifferentiated stage, is characterized by a small network of capillaries. • Stage 2, the retiform stage, involves the development of plexiform structures, increasing the vascular system’s volume and extent. • Stage 3, the maturation stage, occurs by the third week of gestation. The first large arteries, veins, and lymphatic ducts are seen. Vascular development is guided by receptors for growth factors. The blood vessels begin to form on the seventeenth day in the extraembryonic regions, with the generation of blood islands in the mesoderm of yolk sac, the embryonic stalk, and chorionic villus. Subsequently, throughout the embryonic disc the vascular network gradually expands for vasculogenesis (the formation of the circulatory system) and angiogenesis (the sprouting of blood vessels from preexisting angioblastic cords), a gradual process. By the twenty-fourth day, the yolk sac is connected to the embryo through the two veins and three vitelline arteries (celiac trunk,

K23346_Neligan_10_Lymph Malform_r2_0143-0162.indd 144

5/26/2015 8:54:58 AM

Chapter 10  Lymphatic Malformations

145

superior mesenteric artery, and inferior mesenteric artery), and the red blood cells begin to circulate. The two allantoic veins and the two allantoic arteries pass through the pedicle, which will become the umbilical cord. Various veins arise from the edge of the body of the embryo. These vessels are called common cardinal veins: • Two anterior cardinal veins from the cephalic endpoint • Two posterior cardinal veins from the tail endpoint • Two common cardinal veins, left and right Initially the venous system is symmetrical, but during the second month of gestation the right component takes over. The inferior vena cava derives from the right vitelline vein, the left and right posterior cardinal veins, and the superior and inferior cardinal veins. The common cardinal veins, the allantoic veins, and the vitelline veins merge into the venous sinus that, incorporated into the right atrium, will include the orifices of the superior and inferior vena cava. The superior vena cava develops from the common cardinal vein (right branch).

Lymphatic System The lymphatic system derives in part from the mesoderm and in part from the mesenchyme, an undifferentiated tissue that retains the ability to transform into another type of tissue, even in adults. The mesenchymal origin is evident in various cells. The reticular cells constitute the framework of the immunocompetent organs, particularly the lymph nodes, and can transform into macrophages. Endothelial cells, which line the inside of the capillaries and lymph vessels, are able to transform into macrophages. The origin of the lymphatic vessels is not entirely understood. They may develop directly from mesodermal protrusions or from endothelium evaginations of the veins in remodeling (lymphatic sacs). These structures are present in embryos measuring 2 cm (the fourth or fifth week of life or approximately 2 weeks after cardiovascular system development). Six primitive lymph sacs form: • Two equal and symmetrical sacs (jugular sacs) positioned cranially at the union of the jugular veins and the anterior cardinal veins • Two tail sacs (iliac sacs) at the union of the iliac veins and posterior cardinal veins • One median sac (retroperitoneal sac) on the posterior abdominal wall, at the root of the mesentery • One sac (cisterna chyli) dorsal to the retroperitoneal sac The sacs are connected by lymphatic channels. Both main channels develop independently and are formed by primitive right and left thoracic ducts. They join caudally with an anastomotic branch, combining the jugular sacs with the cisterna chyli. In adults, the thoracic duct grows caudally from the distal portions of the primitive thoracic ducts (right and left) and cranially from left thoracic duct. The right lymphatic duct (large lymphatic right vein) derives from the cranial part of the right thoracic duct. The subclavian, jugular, and bronchomediastinal collecting trunks derive from the jugular sacs.

K23346_Neligan_10_Lymph Malform_r2_0143-0162.indd 145

5/26/2015 8:54:58 AM

146

Part II  Anatomy, Physiology, and Lymphangiogenesis

Classification of Vascular Malformations Vascular malformations have multiple causes and complex clinical presentations, making their classification challenging. Malan and Puglionisi6 tried to distinguish them by their anatomy and the presence or absence of arteriovenous fistulas. Szilagyi et al7 divided them into three stages according to the three main phases of embryologic life (segmentation, gastrulation, and organogenesis). According to this classification, capillary malformations (hemangiomatous malformations) occur when development is blocked during the first embryologic phase. (These should not be confused with hemangiomas, which are skin tumors.) Microarteriovenous or macroarteriovenous fistulas occur when development is interrupted during the second embryologic phase. Persistent embryonic veins indicate a developmental block in the third embryologic phase and involve mixed segmental vascular malformations. Jackson et al12 based their classification on hemodynamic and angiography findings. High- or lowflow lesions are distinguished by the amount of blood flow in arteriovenous fistulas. High-flow malformations correspond to the presence of arteriovenous macrofistulas, and low-flow malformations correspond to arteriovenous microfistulas, which carry a better prognosis. The most recent classification was published in Hamburg in 1990.13 Rutherford et al14 and Lee et al15 later revised this classification (Table 10-1). Malformations are classified based on the predominant basic defect as follows: • Arterial • Venous • Lymphatic • Mixed, either truncal or extratruncal forms according to the flow along the main axis or branches Predominantly arterial truncal malformations include congenital forms of sciatic artery or accessory subclavian artery persistence (which, passing behind the esophagus, is responsible for dysphagia lusoria); aortic arch anomalies; coarctation of the thoracic or abdominal aorta; and persistent embryonic vessels. Predominantly venous truncal malformations include forms of persistent embryonic veins, such as the outer marginal vein. These present with mixed forms of aplasia, ectasia, aneurysm, and thrombosis of various anatomic segments. The most frequent alteration is ectasia of the venous system, which occurs in patients with Klippel-Trenaunay syndrome and makes up approximately 40% of the malformations. Venous aneurysms or hypoplasia of the venous wall is much less common, accounting for less than 10% of malformations. Predominantly arteriovenous forms are most frequently extratruncal and may be localized or diffuse, especially on the skin or mucosa. They can be isolated or associated with more complex pathologic syndromes. Low-flow and high-flow shunt malformations can be distinguished. The predominantly arteriovenous malformations are more frequently seen in the lower limbs and pelvis, and the inherited forms usually occur in the lungs and sometimes in the brain. Hereditary hemorrhagic telangiectasia, for example, is characterized by the presence of spider veins in the dermis, mucosa, and viscera and is associated with arteriovenous fistulas in the lungs and brain.16

K23346_Neligan_10_Lymph Malform_r2_0143-0162.indd 146

5/26/2015 8:54:58 AM

147

Chapter 10  Lymphatic Malformations

TABLE 10-1  Hamburg Classification of Congenital Vascular Malformations (Modified) Affected Segment

Anatomic Form

Type

Arterial malformations

Truncal Extratruncal

Aplasia or obstruction Infiltrating Limited

Venous malformations

Truncal

Aplasia or obstruction Dilation Infiltrating Limited

Extratruncal Arteriovenous (arteriovenous shunt malformations)

Truncal Extratruncal

Lymphatic malformations

Truncal Extratruncal

Mixed vascular malformations

Truncal Extratruncal

Deep arteriovenous fistula Superficial arteriovenous fistula Infiltrating Limited Aplasia or obstruction Dilation Infiltrating Limited Arterial and venous Hemolymphatic Infiltrating hemolymphatic Limited hemolymphatic

These are manifested by recurrent nosebleeds, which occur in 90% of patients when they blow their nose or during a race or emotional stress. The mortality rate is 4%.

Capillary Malformations Capillary malformations are also known as hemangiomas. They are most often found on the head and limbs (upper and lower). The dimensions are extremely variable, from localized forms to giant forms with involvement of an entire side of the body. On clinical examination, capillary malformations appear as a red or rosy-purple skin rash with jagged or sharp margins. They are generally not raised, are highly variable in size and scope, and can be isolated or multiple and confluent. Within the patch, telangiectatic striae formed by largercaliber capillaries are evident. In capillary-venous mixed forms, abnormal reticular veins that drain from the malformation can be seen.17,18 Usually the skin lesion is sharply localized to the right or left of the midline, sometimes extending slightly beyond it. Another characteristic clinical aspect is metamerization (that is, the topography of the lesions usually follows the distribution of the head, trunk, or limb dermatomes). Facial locations typically respect the distribution of the sensory branches of the trigeminal nerve. In some patients, especially those with craniofacial lesions, hemangiomas tend to grow and produce a marked hyperplasia of the dermis and subcutaneous tissue. They are associated with a characteristic dilation of the subepidermal venous bed. These lesions are called hyperplastic birthmarks, commonly referred to as nevus flammeus or port-wine stains. On clinical examination, they pre­

K23346_Neligan_10_Lymph Malform_r2_0143-0162.indd 147

5/26/2015 8:54:58 AM

148

Part II  Anatomy, Physiology, and Lymphangiogenesis

sent as patches of very dark purplish-red fibrotic tissue, often covered by vegetating plaques or polypoid formations.19

Lymphatic Malformations Congenital malformations of the lymphatic system are characterized by embryogenetic capillary abnormalities or by malformations of the major lymphatic collectors of the limbs, head, and torso.20 They are more common peripherally, especially in the legs, but can occur in the cervicofacial, thoracic, and pelvic regions.21 According to the Hamburg classification, congenital malformations of the lymphatic system can be distinguished by tissue forms (extratruncal) and cysts forms (truncal forms or lymphangiomas).22 Tissue lymphangiomas (or cystic hygromas) consist of a dense network of lymphatic microscopic vessels. Their dimensions are extremely variable, ranging from a small nodule to a voluminous mass. Lymphangiomas are characterized by an abnormal ectasia with saccular dilation of large lymph collectors or tanks. The locations correspond to the onset of major lymph node stations: submandibular, lateral cervical, axillary, inguinal, and mediastinal. The clinical development of lymphatic malformations is closely related to the type and severity of anatomic abnormalities and the body region affected. The tissue lymphangiomas manifest as skin rashes or subcutaneous swellings with a whitish, warty, and irregular surface, often covered by translucent microvesicles containing serous fluid. Cystic hygromas occur as massive swellings under the skin and have a soft and spongy texture. They float and expand with moderate antigravity maneuvers and are nonpulsating. The behavior of lymphatic malformations is extremely variable. They are usually present at birth and tend to gradually increase over the years, with remissions and relapses influenced by various factors (hormones, trauma, and infection). In some cases such as hemangiomas, progressive involution occurs after puberty.23,24 Lymphatic malformations may manifest clinically with lymphedema, an edema with a particularly high-protein concentration that can lead to permanent disability. Lymphedemas are truncal lymphatic malformations, because they appear after an interruption in the anatomy of a lymphatic region. Truncal lymphatic malformations can develop at birth or later. Lymphedemas are classified as primary or secondary. Primary lymphedema is connatal, early and late, and is often familial or inherited. Secondary lymphedema occurs after surgery, radiotherapy, trauma, and/or inflammation, or is functional (for example, postphlebitic). Secondary lymphedema is increasing in the average population because of the growing incidence of tumors. Treatments for tumors have improved; however, survival is associated with treatment sequelae, mainly secondary lymphedema. Lymphodynamic impairment results from agenesis or aplasia of lymph nodes or lymphatic trunks, impaired permeability of lymph capillaries, or lymphadenodysplasia. It can be familial or inherited (for “sporadic mutations” in only one member of the family) or caused by chromosomal abnormalities, amniotic bands, or other congenital diseases. Prevention is essential. Therefore if familial, it is important to know the genetic mutation in order to study the same mutation in subjects with a blood relation with the proband who has the mutation.

K23346_Neligan_10_Lymph Malform_r2_0143-0162.indd 148

5/26/2015 8:54:58 AM

149

Chapter 10  Lymphatic Malformations

The most frequent complications are usually local. Tissue forms can cause skin or mucosa necrosis, possibly associated with a lymphorrhage. Cystic forms can result in bleeding, infection, and compression of vital organs. More than 70% of vascular malformations are mixed.25 The most common forms seen clinically are listed in Table 10-2.

TABLE 10-2  Complex Vascular Malformations Syndrome

Genetic Description

Blue rubber bleb nevus

Type

Location

Characteristic Features

Autosomal dominant

Cavernous venous malformation

Skin, gastrointestinal tract, spleen, liver, central nervous system

Bluish, compressible rubbery lesions Gastrointestinal bleeding Anemia

Gorham Stout disease

Not hereditary

Angiomas Multiple lymphatic and venous malformations

Bones: shoulder, skull, pelvic girdle, jaw, ribs, and spine

Uncontrolled proliferation of thinwalled vascular or lymphatic vessels within bone, which leads to resorption and replacement of bone with angiomas and/or fibrosis

Hennekam syndrome

Autosomal recessive

Multiple lymphatic malformations

Viscera, bones, pulmonary system, face

Lymphedema Anomalies of the teeth, gingival hypertrophy Seizures Vascular anomalies Congenital pulmonary lymphangiec­ tasia Narrowness of the upper chest

Kasabach-Merritt syndrome

Autosomal dominant

Large cavernous venous malformation

Trunk, extremities

Thrombocytopenia Hemorrhage Anemia Ecchymosis Purpura

Klippel-Trenaunay syndrome

Somatic mutations

Few or low-flow arteriovenous shunt Venous or lymphatic trunk malformations Port-wine stains

Extremities, pelvis

Soft tissue and bone hypertrophy Varicosities (lateral lower limb to foot) Capillary or venous-lymphatic malformations

Maffucci syndrome (dyschondroplasia with vascular hamartoma)

Probably autosomal dominant

Arteriovenous malformations Cavernous lymphangioma

Fingers, toes, extremities, viscera

Enchondromas Spontaneous fractures Short extremity Vitiligo

Noonan syndrome

Autosomal dominant

Lymphatic or venouslymphatic malformations

Face, heart, skin, lower limbs, nervous system

Unusual facies (hypertelorism, downward-slanting eyes, and webbed neck) Congenital heart disease Short status Chest deformity Mental retardation Bleeding diathesis

Continued

K23349_Neligan_10_Lymph Malform_r3_0143-0162.indd 149

5/28/2015 8:16:23 AM

150

Part II  Anatomy, Physiology, and Lymphangiogenesis

TABLE 10-2  Complex Vascular Malformations—cont’d Syndrome

Genetic Description

Parkes-Weber syndrome

Type

Location

Characteristic Features

Somatic mutations

Arteriovenous malformations (intraosseous or close to epiphyseal plate) Port-wine stain

Extremities, pelvis

Soft tissue and bone hypertrophy Atypical varicosity Capillary arteriovenous shunts High-flow arteriovenous shunts

Proteus syndrome

Somatic dominant gene mutation (lethal, except with mosaicism)

Multicutaneous and visceral arteriovenous, lymphatic, and venous malformations with muscle and skeletal diseases

Fingers, toes, extremities, viscera

Soft tissue abnormalities (lipomatosis, fatty hypertrophy, regional fatty atrophy, hyperpigmentation or cutaneous nevi, hyperkeratosis of the palms and soles, macrodactyly of the hands and feet, exostosis, bilateral genu valgum, kyphosis, and scoliosis secondary to vertebral dysplasia

Rendu-OslerWeber syndrome (h­ereditary hemorrhagic telangiectasia)

Autosomal dominant

Punctate angioma Telangiectasia Arteriovenous malformation of the gastrointestinal tract

Skin, mucous membranes, liver, lungs, kidneys, brain, spinal cord

Epistaxis Hematemesis, melena Hematuria Hepatomegaly Neurologic symptoms

Riley-Smith syndrome

Autosomal dominant

Multiple hemangiomas

Skin, brain, gastrointestinal tract, genitalia, thyroid

Symmetrical macrocephaly Pigmented maculae of the glans penis Mesodermal hamartomas (primarily subcutaneous and visceral lipomas, multiple hemangiomas, and intestinal polyps) Dysmorphy

Servelle-Martorell syndrome

Venous and rarely arterial malformations Cavernous hemangiomas

Limbs

Limb undergrowth, capillary stains, and varicose veins

Sturge-Weber syndrome (encephalotrigeminal angiomatosis)

Port-wine stains

Trigeminal area, leptomeninges, choroid oral mucosa

Convulsions Hemiplegia Ocular deformities Mental retardation Glaucoma Intracerebral calcifications

Hemangiomas

Retina, cerebellum

Cysts in cerebellum, pancreas, liver, adrenals, and kidneys

von HippelLindau (oculocerebellar hemangioblastomatosis)

Autosomal dominant

K23346_Neligan_10_Lymph Malform_r2_0143-0162.indd 150

5/26/2015 8:54:58 AM

Chapter 10  Lymphatic Malformations

151

Genetics of Vascular Receptors During embryonic development, vasculogenesis is mediated by growth factors (produced by the germ layers) and their receptor sites on the endothelial cells of the vascular system (see Chapter 4). In adults, growth factors are produced to a lesser extent. Their role in the regulation of angiogenesis is under study. Models that can help in understanding the mechanisms of suppression, control, and expression are the neoplastic cells, which, both in vitro and in vivo, reproduce the events that lead to the differentiation from mesodermal cells to the final cells. Among the growth regulators and their receptors, the most studied are vascular endothelial growth factor/vascular endothelial growth factor receptors (VEGF/VEGFR), forkhead box protein C2 (FOXC2), and SRY-related HMG box 18 (SOX18). Some genetic disorders currently under study in tumor development in animal models have shown a close relationship with vascular malformations. Some proteins are known growth factors such as hepatocyte growth factor (HGF); some genes encode junction proteins, which reshape cellular connections and can change the microvacular tissue unit function of absorption (see Table 10-3).

The Role of VEGF-3: Gene Map Locus 5q34.35 Vascular growth factors are important signaling proteins involved in vasculogenesis and angiogenesis. The activity of VEGF is mainly targeted to vascular endothelial cells, but also has effects on other types of cells (for example, monocytes and macrophages). In vitro VEGFs have proved capable of stimulating mitosis of endothelial cells and cell migration. VEGFs also increase microvascular permeability and are sometimes referred to as vascular permeability factors. All members of the VEGF family stimulate cellular responses by binding to receptor tyrosine kinases (VEGFR-X) on the surface of cells, causing their dimerization and activation through transphosphorylation. The receptors of VEGF have an extracellular portion composed of seven immunoglobulin domains, a single transmembrane region, and an intracellular portion containing tyrosine kinase. The most well-known growth factor, and the first to be isolated from the blood capillaries, is VEGF-A, which binds to two receptors: VEGFR-1 and VEGFR-2. Recently, a new receptor (VEGFR-3) was isolated. It does not bind VEGF-A. Instead, it binds two other growth factors, VEGF-C and VEGF-D, which are mainly expressed on lymphatic capillaries.26,27 He et al28 made an important contribution to this field by studying the behavior of cancer cells and identifying VEGF-C, a specific growth factor for lymphatic tissue that promotes lymphangiogenesis. VEGF-C is produced from a variety of human tumors, and its deregulation induces lymphangiogenesis and promotes tumor metastasis through lymphatics. Lymphangiogenesis induced by VEGF-C is primarily determined by the activation of tyrosine kinase bound to the receptor VEGFR-3. VEGFR-3 is expressed almost exclusively on lymphatic endothelium in normal adults. However, in studies with guinea pigs, VEGFR-3 is widely expressed on endothelial cells of the blood system, before lymphatic vessels appear in the fourth week of gestation.

K23346_Neligan_10_Lymph Malform_r2_0143-0162.indd 151

5/26/2015 8:54:58 AM

152

Part II  Anatomy, Physiology, and Lymphangiogenesis

VEGF-D is another growth factor that activates VEGFR-3; its overexpression leads to accelerated lymphangiogenesis and the spread of cancer cells to lymph nodes. In turn, the inactivation of VEGF-C and VEGF-D inhibits tumor lymphangiogenesis. Unlike blood capillaries, lymphatic capillaries are lined by a single layer of nonfenestrated endothelial cells that lack pericytes, smooth muscle cells, and a basal membrane. VEGFR-3 is also expressed on the blood capillaries of normal breast tissue, on chronic wounds, and on neuroendocrine organs29,30; monocytes, macrophages, and some dendritic cells express this receptor. The receptor VEGFR-2, which is the main promoter of mitosis of endothelial cells in the blood and binds the factors VEGF-A, VEGF-C, and VEGF-D, is also expressed on lymphatic capillaries and mediates lymphangiogenesis. Lymphangiogenesis was prevented through the selective blockade of VEGFR-3, using an antibody (MF4-31C1) in a murine model for the study of regeneration lymphatics.31 Even in the presence of an overexpression of VEGF-C in breast cancer cells, the antibody MF4-31C1 reduced lymphangiogenesis and lymphatic metastases, confirming the essential role of this receptor. The preexisting lymphatic vessels did not appear morphologically or functionally altered by a prolonged blockade of VEGFR-3, and blood vessels, including those resulting from neovascularization, were only slightly reduced in number. Another potential target of a VEGFR-3 block may be the immune system, leading to disorders such as inflammatory bowel disease. In ulcerative colitis and Crohn disease, the number of lymphatic capillaries is substantially increased in the lamina propria and submucosa of the small and large intestine, which is thought to contribute to the morbidity of the disease.32 The preexisting lymphatic vessels did not appear to be substantially modified by the blockade of VEFGR-3. However, further in vitro studies have revealed the importance of VEGF-C/VEGFR-3 in the survival of lymphoid cells.33 The expression of VEGFR-3 increases in blood capillaries of chronic wounds, although the prolonged inhibition of VEGFR-3 only minimally influences the regeneration of capillaries. Mediation by interaction with VEFGR-2 probably plays an important role in vivo, along with the expression of VEGF-D, which binds to and activates VEGFR-3 and VEGFR-2.

The Role of FOXC2: Gene Map Locus 16q24.3 FOXC2 is a protein that in humans is encoded by the gene FOXC2, part of the family of transcription factors. FOXC2, similar to VEGF, is involved in the promotion of metastasis by lymph nodes. In particular, the expression of FOXC2 increases when the epithelial cells are subjected to a mesenchymalepithelial transition. The repression of FOXC2, using shRNA, reduces the metastatic ability of breast cancer cells.34 Mutations in the FOXC2 gene have been associated with multiple lymphedematous syndromes, including lymphedema-distichiasis syndrome, lymphedema–yellow nail syndrome, and

K23346_Neligan_10_Lymph Malform_r2_0143-0162.indd 152

5/26/2015 8:54:58 AM

Chapter 10  Lymphatic Malformations

153

lymphedema-ptosis, and they play an important role in the genesis of varicose veins. In addition, some FOXC2 mutations have been identified in patients with family obesity, diabetes mellitus, and adrenal insufficiency, especially in European families. FOXC2 is an essential transcription factor of the cardiovascular system and its diseases. However, the cellular and molecular functions of FOXC2 in vascular endothelial cells have not been fully understood. Some authors have identified targets of FOXC2, including molecules associated with cellular-extracellular matrix: integrin beta-3 (ITGB3), integrin beta-5 (ITGB5), and fibronectin. In particular, the expression of ITGB3 is directly regulated by FOXC2. In vitro, ITGB3 is a known regulator of angiogenesis. Overexpression of FOXC2 greatly improves the adhesion of endothelial cells, and this effect is strongly inhibited by neutralization, by antibodies of ITGB3. In addition, overexpression of FOXC2 enhances the tropism of the small vessels and intensifies angiogenesis.35 The normal lymphatic capillaries are devoid of smooth muscle cells, whereas the lymphatic manifolds are surrounded by a layer of smooth muscle cells, which allows the progression of the lymph in a centripetal direction, aided by the presence of one-way valves. The lymphatic capillaries of the lower limbs of patients who have mutations in FOXC2 are surrounded by smooth muscle cells abnormally organized, and lymphatic collectors are almost entirely lacking. In individuals without mutations in FOXC2, only the capillaries are surrounded by smooth muscle cells; however, in those with mutations in FOXC2, the lymphatic capillaries are surrounded by smooth, disorganized muscle cells.

The Role of SOX18: Gene Map Locus 20q13.33 The SOX18 transcription factor genes (sex determining region Y [SRY] and high mobility group [HMG] box) are a family of DNA-binding domain protein through the HMG. They play a key role in the embryonic development of eukaryotic organisms and in the regulation of gene expression required for cell differentiation. The transcription factor SOX18 is recognized as a specific switch in inducing the differentiation of endothelial cells of the blood vascular system to express prospero-related homeobox 1 (PROX1), a gene critical for the formation of lymphatic vessels from the cardinal veins. In experimental models, SOX proteins are transcription factors that regulate normal cell function through links on specific DNA sites that allow/inhibit the control of certain genes in the cells of the lungs, musculoskeletal system, cardiovascular system,36 brain, testes, liver, and leukocytes. Some studies, always on animal models, have demonstrated that the expression of SOX genes is present both during embryonic life and adult life.37 In experimental models of cancer progression, the selective blockade of the genes coding for the proteins of SOX18 transcription leads to 80% reduction of tumor angiogenesis. However, the selective blockade in experimental adult mice models leads to the development of cardiovascular diseases. Even in humans the transcriptional proteins family -SOX (SRY-related and HMG-box) are both involved in embryonic development and in the cellular regulation of adult life. The protein encoded by the gene, after forming a protein complex with other cytoplasmic proteins, regulates function in cells of blood vessels, lymphatic vessels, and hair. Mutations in this gene have been associated with recessive and dominant forms of hypotrichosis-lymphedema-telangiectasia syndrome (Table 10-3).

K23346_Neligan_10_Lymph Malform_r2_0143-0162.indd 153

5/26/2015 8:54:59 AM

154

Part II  Anatomy, Physiology, and Lymphangiogenesis

TABLE 10-3  Genetic Alterations Involved in Vascular Disorders Currently Under Study Gene

GeneCards Description

Hepatocyte growth factor (HGF)

HGF regulates cell growth, cell motility, and morphogenesis by activating a tyrosine kinase signaling cascade after binding to the protooncogenic c-Met receptor. Its ability to stimulate mitogenesis, cell motility, and matrix invasion confers a central role in angiogenesis, tumorogenesis, and tissue regeneration. Diseases associated with HGF include dfnb39 nonsyndromic hearing loss and deafness and autosomal recessive deafness.

Mesenchymal epithelial transition factor (MET)

MET, also known as hepatocyte growth factor receptor (HGFR), is a protooncogenic receptor tyrosine kinase. MET receptors are expressed on cells of epithelial origin. Aberrant activation of the HGF/MET pathway leads to a variety of cancers. MET mutation is associated with a poor prognosis, because it can trigger tumor growth, angiogenesis, and metastasis. Diseases associated with MET include papillary renal carcinoma and papillary carcinoma.

Gap junction protein gamma 2 (GJC2)

The GJC2 gene encodes a gap junction protein. Defects in this gene are the cause of autosomal recessive Pelizaeus-Merzbacher-like disease. Gap channels (also known as gap junctions) are specialized cell-cell contacts between almost all eukaryotic cells that provide direct intracellular communication. They are continuously synthesized and degraded, allowing tissues to rapidly adapt to changing environmental conditions. Connexins play a key role in many physiologic processes, including cardiac and smooth muscle contraction, regulation of neuronal excitability, epithelial electrolyte transport, and keratinocyte differentiation. Mutations in connexin genes are associated with human diseases, including sensorineural deafness, a variety of skin disorders, peripheral neuropathy, vascular abnormalities, and cardiac disease. Diseases associated with GJC2 include hereditary lymphedema 1C, and spastic paraplegia.

GATA binding protein (GATA-2)

GATA-2 encodes a member of the GATA family of zinc-finger transcription factors that are named for the consensus nucleotide sequence they bind in the promoter regions of target genes. The encoded protein plays an essential role in regulating the transcription of genes involved in the development and proliferation of hematopoietic and endocrine cell lineages. Diseases associated with GATA-2 include myelodysplastic syndromes and acute myeloid leukemia, susceptibility, gata2-related. Related superpathways are DREAM repression and dynorphin expression and platelet activation, signaling, and aggregation.

Vascular cell adhesion molecule (VCAM-1)

VCAM-1 is a member of the Ig superfamily and encodes a cell surface sialoglycoprotein expressed by cytokine-activated endothelium. This type I membrane protein mediates leukocyte–endothelial cell adhesion and signal transduction, and may play a role in the development of artherosclerosis and rheumatoid arthritis. Diseases associated with VCAM-1 include Mooren ulcer and vasculitis.

Fatty acid binding protein (FABP4)

FABP4 encodes the fatty acid binding protein found in adipocytes. Fatty acid binding proteins are a family of small, highly conserved, cytoplasmic proteins that bind long-chain fatty acids and other hydrophobic ligands. They bind long-chain fatty acids and retinoic acid and deliver long-chain fatty acids and retinoic acid to their cognate receptors in the nucleus. Diseases associated with FABP4 include back pain and bladder transitional cell carcinoma.

K23346_Neligan_10_Lymph Malform_r2_0143-0162.indd 154

5/26/2015 8:54:59 AM

Chapter 10  Lymphatic Malformations

155

Diagnosis Ultrasonography Doppler ultrasonography is an excellent tool for the initial assessment of patients with venous malformations. High- and low-flow forms are easily distinguished. High-flow malformations show waves with high speed and low resistance; in low-flow malformations, the volumes, arterial flow, and arterial resistance are normal in the arteries that supply the venous side. Flows with low resistance can be found in intramuscular malformations. Ultrasonography provides additional information on the morphology and evolution of venolymphatic lesions. The malformed veins are more or less compressible, depending on the parietal thickening that occurs with time. Patients can be assessed for venous thrombotic events. In those undergoing surgical or endovascular treatment, the incidence of deep venous thrombosis is high, beginning on postoperative day 1.38

Magnetic Resonance Imaging and Computed Tomography MRI provides information on only high- and low-flow forms, but also on the closer organs and surrounding tissues (muscles, bones, nerves, and adipose tissue). The venolymphatic malformations (low-flow) show a high signal intensity in the long TR/TE sequences, whereas the arterial malformations (high-flow) do not show an increase in the signal. Furthermore, the venous-lymphatic malformations give a signal intensity higher than that of muscle or skeleton in the T1- and T2-weighted images, probably because of the presence of a fibrous septum along the vascular endothelium. MRI, compared with arteriography or venography, allows better assessment of the extent of the malformation, especially in cases in which access to venous contrast fluid is difficult.39 Two-dimensional high-resolution CT with three-dimensional reconstructions provides good information on the extent of malformation, but does not distinguish between lesions that are predominantly arterial and mixed lesions. Because of the need for contrast, timing is important. (It is an operator-dependent examination.) Proper measurement of the lesion depends on the amount of blood that it contains.40 CT provides important information about the tissue characteristics of the suprafascial and subfascial compartments. It is helpful for developing a therapeutic approach and for monitoring the results of treatment.

Arteriography and Venography Arteriography is reserved for patients who will undergo embolization. Angiography is useful for measuring the size of the nurse artery and assessing the extent of the shunt. The flow volume is calculated from the size and proportion of the source artery opacification, whereas the shunt volume can be roughly calculated using the time of the appearance of contrast in the veins.41 Venography is reserved for patients who will undergo venous correction. The prevalence of venous malformations in the nurse artery is not increased, because the size of the capillary bed maintains a low resistance. Arteriography does not correctly show the extent of the malformation from the venous side. In these cases, injection of the contrast in the venous side reveals the complete vascular malformation.42

K23346_Neligan_10_Lymph Malform_r2_0143-0162.indd 155

5/26/2015 8:54:59 AM

156

Part II  Anatomy, Physiology, and Lymphangiogenesis

Lymphoscintigraphy Lymphoscintigraphy is the benchmark of diagnostic imaging in patients with primary and secondary lymphedema. In the current guidelines, however, the method is not yet standardized, reducing the specificity of the examination. Lymphoscintigraphy had proved useful for identifying and defining lymphedemas; in directing preventive, physical, and surgical treatments; and in determining a prognosis. In patients with subclinical lymphedema, the appearance of lymph nodes along the limb (particularly after a lymphadenectomy) is interpreted as a sign of lymph transport failure. The examination provides morphologic and functional information about lymphatic circulation; a sterile technique is essential for accurate interpretation of data. Morphology demonstrated by lymphoscintigraphy includes the following: • Hypogenesis or agenesis of lymphatic stations or one-way lymphatic vessels or valves • The presence and extent of dermal backflow • The presence of lymph node obstructions in the middle of the limb (that is, the knee and the elbow) • The presence of alternative channels

Treatment There are absolute and relative indications for the treatment of vascular malformations. Absolute indications include the following: • Hemorrhage • Ischemia • Ulcers refractory to conventional treatments • Congestive heart failure Clinical manifestations for which treatment is absolutely indicated include the following: • Intramuscular or retroperitoneal hematoma • Hematuria • Rectal bleeding • Hematemesis • Hemoptysis • Unifocal/multifocal bleeding in the brain or spinal cord Relative indications for treatment are the following: • Pain • Intermittent claudication • Leg length discrepancy • Other aesthetic alterations A multidisciplinary approach to the evaluation and treatment of vascular malformation provides the best outcome, especially for young patients.

K23346_Neligan_10_Lymph Malform_r2_0143-0162.indd 156

5/26/2015 8:54:59 AM

Chapter 10  Lymphatic Malformations

157

Conservative treatment is recommended for most patients. Minimally invasive percutaneous techniques have evolved in recent years and include scleroembolization, a very complex procedure. Laser therapy has been used effectively for capillary malformations (port-wine stains and other imperfections). Surgical resection is reserved for patients who have superficial localized lesions or bulky varicose veins that developed when the patient was young and are debilitating. Compression is the most important element43 in the management of primary and secondary lymphedema.44,45 After the affected limb or limbs have undergone anatomic changes, bandages are placed to maintain constant external pressure.46 According to the Young-Laplace equation,47 compression, at constant tension, is directly dependent on the radius of the affected limb: Compression 5 Layers of bandages 3

Bandage tension Leg radius

A recent study showed that compression is most effective when patient compliance with the bandaging requirements is high.48 The bandage should be as comfortable as possible to ensure that it is kept in place and allows the most effective drainage.49 Lymphatic drainage depends on the level of injury of the lymphatic system and its residual function, which is influenced by the mobility of the limb. The patient’s lifestyle is a factor in the effectiveness of treatment. Good compliance with instructions for care of the bandage and the overlying elastic garment are essential. Venolymphatic malformations comprise a series of various-sized cysts covering the lymphatic vascular endothelium. Cysts in serum or whole blood may or may not communicate with each other and are immersed in a support matrix consisting of fibrous tissue, smooth muscle cells, and lymphocytic aggregates. It is possible to distinguish macrocystic lesions (cystic hygroma) from microcystic and mixed lesions. Surgical resection is considered the standard treatment, although the recurrence rate varies from 15% to 53%,50 and significant complications (nerve palsy) occur in up to a third of cases. Recurrence of cystic hygroma and serious surgical complications can be prevented with the use of interventional radiologic techniques. Minimally invasive radiotherapy50,51 is helpful for ablating selective vascular endothelial multicystic lesions52 through the use of different agents, including bleomycin, doxycycline, ethanol, Ethibloc, OK-432, and sodium tetradecyl sulfate. Therapeutic success is reported to vary from 20% to 64%. Macrocystic lesions (larger than 1 cm in diameter) are catheterized percutaneously under ultrasonographic guidance along different points of the malformation and then are injected with sclerosing agents. The best results, with a therapeutic success rate of 80% to 90%, are obtained with doxycyline, OK-432, and bleomycin.53-55 Minor complications such as hemolytic anemia, hypoglycemia, metabolic acidosis, transient hypotension, blisters, and hair loss develop in 10% to 14% of cases.56,57

K23346_Neligan_10_Lymph Malform_r2_0143-0162.indd 157

5/26/2015 8:54:59 AM

158

Part II  Anatomy, Physiology, and Lymphangiogenesis

Microcystic malformations are treated with a single percutaneous injection under ultrasound guidance. Cysts are aspirated and sclerosing foam (usually doxycycline) is injected, changing the microbial environment of the treated areas. The therapeutic success rate is more than 90%.58 Surgical treatment is reserved for more complex forms of vascular malformations, especially those with low flow, and to compose venous flow in prevailing venous component. Surgery is preceded and/or followed by sclerotherapy of large areas of the lesion. The malformed component is usually excised in a bloodless field, and an Esmarch bandage is placed.59 In high-flow forms, the only necessary surgical treatment is amputation: amputation is often a life-saving medical treatment, because it corrects hemodynamic decompensation created by the shunt.60

Conclusion The approach to patients with vascular malformations is multidisciplinary, requiring a team that performs medical, radiologic, and surgical procedures and provides rehabilitation and psychological care. Correct assessment and the study of disease evolution are the first and most difficult steps in understanding and managing patients with vascular malformations. The diagnosis and therapy for patients with vascular malformations is extremely challenging. The clinical features are variable, from small cutaneous manifestations to complex deformity of organs and limbs. Initial assessments are performed to study the nature of the malformation, including its size and complexity, and to quantify the damage caused by hemodynamic alterations. The method chosen must be repeated over time to monitor the evolution of the lesion and the success of therapy. Ultrasonography is relatively inexpensive and clearly defines the hemodynamic profile of the malformation, especially in venolymphatic (low-flow) lesions. It is useful in the early stages of disease and can be a good corollary examination in more complex evaluations. Ultrasound examinations are operator dependent; diagnostic accuracy is strongly affected by experience. MRI (with or without a contrast medium) is perhaps the most comprehensive investigation for determining the morphofunctional damage. This mode is not strictly linked to the experience of the operator for diagnostic accuracy and is easily comparable and repeatable over time. Arteriography and phlebography are instrumental investigations carried out if contextual treatment is needed or if the vascular malformation is complex and the therapeutic choice difficult. Nonlethal vascular malformations can be disabling, affecting a patient’s a social life and self-image. They can influence quality of life by impairing the individual’s physical performance and through chronic clinical worsening. Complex surgical therapy is performed in a few specialized centers for patients with unfavorable results of hemodynamic assessment. Rehabilitation therapy, including medication and physical therapy, slows progression of the disease, allowing patients to be more autonomous in managing their activities of daily life, thus enhancing their quality of life. Furthermore, medical and physical therapy may be carried out more easily on the affected area, costs are lower, and optimal timing of surgery (corrective and demolitive) can be chosen.

K23346_Neligan_10_Lymph Malform_r2_0143-0162.indd 158

5/26/2015 8:54:59 AM

Chapter 10  Lymphatic Malformations

159

R EFERENCES 1. Eifert S, Villavicencio L, Kao TG, et al. Prevalence of deep venous anomalies in congenital vascular malformations of venous predominance. J Vasc Surg 31:462-471, 2000. 2. Servelle M. Klippel and Trénaunay’s syndrome: 768 operated cases. Ann Surg 201:365-373, 1985. 3. Virchow R. Pathologie des Tumeurs: Cours Professé à l’Université de Berlin, vol 4. Paris: GermerBaillière, 1876. 4. Nicoladoni C. Phlebarteriectasie der rechten oberen Extremität. Arch Klin Chir 18:252-274, 1875. 5. Branham HH. Aneurysmal varix of the femoral artery and vein following a gunshot wound. Int J Surg 3:250-255, 1890. 6. Malan E, Puglionisi A. Congenital angiodysplasias of the extremities: generalities and classifications: venous dysplasias. J Cardiovasc Surg 5:87-130, 1964. 7. Szilagyi DE, Smith RF, Elliott JP, et al. Congenital arteriovenous anomalies of the limbs. Arch Surg 111:423-429, 1976. 8. Mulliken JB, Glowacki J. Hemangiomas and vascular malformations in infants and children: a classification based on endothelial characteristics. Plast Reconstr Surg 69:412-420, 1982. 9. Mulliken JB, Zetter BR, Folkman J. In vitro characteristics of endothelium from hemangiomas and vascular malformations. Surgery 92:348-353, 1982. 10. Glowacki J, Mulliken JB. Mast cells in hemangiomas and vascular malformations. Pediatrics 70:48-51, 1982. 11. Woollard HH. The development of the principal arterial stems in the forelimb of the pig. Contrib Embryol 14:139-154, 1922. 12. Jackson IT, Forbes G, May GT. Vascular anomalies. In Mustarde J, Jackson IT, eds. Plastic Surgery in Infancy and Childhood, ed 3. London: Churchill Livingstone, 1988. 13. Belov ST. Classification of congenital vascular defects. Int Angiol 9:141-146, 1990. 14. Rutherford RB, Anderson BO, Durham JD. Congenital vascular malformations of the extremities. In Moore WS, ed. Vascular Surgery: A Comprehensive Review, ed 5. Philadelphia: WB Saunders, 1998. 15. Lee BB, Do YS, Yakes W, et al. Management of arteriovenous malformations: a multidisciplinary approach. J Vasc Surg 39:590-600, 2004. 16. Guttmacher AE, Marchuk DA, White RI. Hereditary hemorrhagic telangiectasia. N Engl J Med 33:918924, 1995. 17. Enjolras O. Angiomas in children at a turning point of understanding and management. Arch Pediatr 6:1261-1265, 1999. 18. Herbreteau D, Brunereau L, Cottier J, et al. Hemangiomas and superficial vascular malformations of the head and neck. Classification, diagnosis, treatment. J Neuroradiol 24:274-290, 1997. 19. Wassef M, Enjorlas O. Superficial vascular malformations: classification and histopathology. Ann Pathol 19:253-264, 1999. 20. Enjorlas O. Classification and management of the various superficial vascular anomalies: hemangiomas and vascular malformations. J Dermatol 24:701-710, 1997. 21. Alqahtani A, Nguyen LT, Flageole H, et al. 25 years’ experience with lymphangiomas in children. J Pediatr Surg 34:1164-1168, 1999. 22. Orvidas LJ, Kasperbauer JL. Pediatric lymphangiomas of the head and neck. Ann Otol Rhinol Laryngol 109:411-421, 2000. 23. Gallagher PG, Mahoney MJ, Gosche JR. Cystic hygroma in the fetus and newborn. Semin Perinatol 23:341-356, 1999. 24. Legiehn GM, Heran MK. Classification, diagnosis, and interventional radiologic management of vascular malformations. Orthop Clin North Am 37:435-474, 2006. 25. Gloviczki AA, Noel AA, Hollier LH. Arteriovenous fistulas and vascular malformations. In Ascher E, ed. Haimovici’s Vascular Surgery, ed 5. Malden, MA: Blackwell, 2004.

K23346_Neligan_10_Lymph Malform_r2_0143-0162.indd 159

5/26/2015 8:54:59 AM

160

Part II  Anatomy, Physiology, and Lymphangiogenesis

26. Saharinen P, Tammela T, Karkkainen MJ, et al. Lymphatic vasculature: development, molecular regulation and role in tumor metastasis and inflammation. Trends Immunol 25:387-395, 2004. 27. Oliver G. Lymphatic vasculature development. Nat Rev Immunol 4:35-45, 2004. 28. He Y, Kozaki K, Karpanen T, et al. Suppression of tumor lymphangiogenesis and lymph node metastasis by blocking vascular endothelial growth factor receptor 3 signaling. J Natl Cancer Inst 94:819-825, 2002. 29. Valtola R, Salven P, Heikkila P, et al. VEGFR-3 and its ligand VEGF-C are associated with angiogenesis in breast cancer. Am J Pathol 154:1381-1390, 1999. 30. Paavonen K, Puolakkainen P, Jussila L, et al. Vascular endothelial growth factor receptor-3 in lymphangiogenesis in wound healing. Am J Pathol 156:1499-1504, 2000. 31. Pytowski B, Goldman J, Persaud K, et al. Complete and specific inhibition of adult lymphatic regeneration by a novel VEGFR-3 neutralizing antibody. J Natl Cancer Inst 97:14-21, 2005. 32. Geleff S, Schoppmann SF, Oberhuber G. Increase in podoplanin-expressing intestinal lymphatic vessels in inflammatory bowel disease. Virchows Arch 442:231-237, 2005. 33. Mäkinen T, Veikkola T, Mustjoki S, et al. Isolated lymphatic endothelial cells transduce growth, survival and migratory signals via the VEGF-C/D receptor VEGFR-3. EMBO J 20:4762-4773, 2001. 34. Mani SA, Yang J, Brooks M, et al. Mesenchyme Forkhead 1 (FOXC2) plays a key role in metastasis and is associated with aggressive basal-like breast cancers. Proc Natl Acad Sci U S A 104:10069-10074, 2007. 35. Hayashi H, Sano H. The FOXC2 transcription factor regulates angiogenesis via induction of integrin beta3 expression. J Biol Chem 283:23791-23800, 2008. 36. Downes M, Koopman P. SOX18 and the transcriptional regulation of blood vessel development. Trends Cardiovasc Med 11:318-324, 2011. 37. Matsui T, Kanai-Azuma M, Hara K, et al. Redundant roles of Sox17 and Sox18 in postnatal angiogenesis in mice. J Cell Sci 119(Pt 17):3513-3526, 2006. 38. Yakes WF, Ray RL, Stavros AT, et al. Diagnostic evaluation by different species of congenital vascular defects. In Balas P, ed. Progress in Angiology. Torino, Italy: Minerva Medica, 1990. 39. Rak KM, Yakes WF, Ray RL, et al. MR imaging of symptomatic peripheral vascular malformations. AJR Am J Roentgenol 159:107-112, 1992. 40. Mulligan PR, Prajapati HJ, Martin LG, et al. Vascular anomalies: classification, imaging characteristics and implications for interventional radiology treatment approaches. Br J Radiol 87(1035):20130392, 2014. 41. Natali J, Merland JJ. Superselective arteriography and therapeutic embolisation for vascular malformation. (Angiodysplasias). J Cardiovasc Surg (Torino) 17:465-472, 1976. 42. Yakes WF. Extremity venous malformation: diagnosis and management. Semin Intervent Radiol 11:332339, 1994. 43. Planinsek Rucigaj T, Tlaker Zunter V, Miljković J. [Compression therapy for lymphedema: our experience] Acta Med Croatica 64:167-173, 2010. 44. Compression therapy. Proceedings of a meeting organized by the International Compression Club. September 2008. Lucca, Italy. Int Angiol 29:391-470, 2010. 45. Linnitt N, Davies R. Fundamentals of compression in the management of lymphoedema. Br J Nurs 16:588-592, 2007. 46. Vaillant L, Müller C, Goussé P. [Treatment of limbs lymphedema] Presse Med 39:1315-1323, 2010. 47. Laplace PS. Essai Philosophique sur les Probabilités. Paris: Courcier, 1814. 48. Lee B, Andrade M, Bergan J, et al; International Union of Phlebology. Diagnosis and treatment of primary lymphedema. Consensus document of the International Union of Phlebology (IUP)-2009. Int Angiol 29:454-470, 2010. 49. Lay-Flurrie K. Use of compression hosiery in chronic oedema and lymphedema. Br J Nurs 20:418, 420, 422, 2011.

K23346_Neligan_10_Lymph Malform_r2_0143-0162.indd 160

5/26/2015 8:54:59 AM

Chapter 10  Lymphatic Malformations

161

50. Zhou Q, Zheng JW, Mai HM, et al. Treatment guidelines of lymphatic malformations of the head and neck. Oral Oncol 47:1105-1109, 2011. 51. Chaudry G, Burrows PE, Padua HM, et al. Sclerotherapy of abdominal lymphatic malformations with doxycycline. J Vasc Interv Radiol 22:1431-1435, 2011. 52. Lowe LH, Marchant TC, Rivard DC, et al. Vascular malformations: classification and terminology the radiologist needs to know. Semin Roentgenol 47:106-117, 2012. 53. Okazaki T, Iwatani S, Yanai T, et al. Treatment of lymphangioma in children: our experience of 128 cases. J Pediatr Surg 42:386-389, 2007. 54. Alomari AI, Karian VE, Lord DJ, et al. Percutaneous sclerotherapy for lymphatic malformations: a retrospective analysis of patient-evaluated improvement. J Vasc Interv Radiol 17:1639-1648, 2006. 55. Lee BB, Kim YW, Seo JM, et al. Current concepts in lymphatic malformation. Vasc Endovascular Surg 39:67-81, 2005. 56. Burrows PE, Mitri RK, Alomari A, et al. Percutaneous sclerotherapy of lymphatic malformations with doxycycline. Lymphat Biol Res 6:209-216, 2008. 57. Cahill AM, Njis E, Ballah D, et al. Percutaneous sclerotherapy in neonatal and infant head and neck lymphatic malformations: a single center experience. J Pediatr Surg 46:2083-2095, 2011. 58. Shiels WE, Dent CM, Murakami JW. Percutaneous treatment of microcystic lymphatic malformation. Presented at the Radiological Society of North America Scientific Assembly and Ninety-sixth Annual Meeting, Chicago, Sept 2010. 59. Villavicencio JL, Gillespie DL, Kreishman P. Controlled ischemia for complex venous surgery: the technique of choice. J Vasc Surg 34:947-951, 2001. 60. Trout HH III, McAllister HA Jr, Giordano JM, et al. Vascular malformations. Surgery 97:36-41, 1985.

K23346_Neligan_10_Lymph Malform_r2_0143-0162.indd 161

5/26/2015 8:54:59 AM

K23346_Neligan_10_Lymph Malform_r2_0143-0162.indd 162

5/26/2015 8:54:59 AM

C hapter 11 Immune Regulation by the Peripheral Lymphatics David G. Hancock

K ey P oints • Lymphedema is characterized by numerous immune deficits, including altered wound healing and increased susceptibility to infection, which develop after an initial lymphatic insult. This connection between immune dysfunction and peripheral lymphatic dysfunction is also mirrored in most human inflammatory disorders, suggesting a crucial immune role for the lymphatics.

Imm

• The peripheral lymphatics are a highly integrated component of the immune system capable of mediating their effector function in response to pathogenic and immunogenic stimuli in the local immune microenvironment. • After activation by a diverse range of stimuli, the lymphatics upregulate a range of immune modulatory molecules capable of mediating cell migration and local immune function. Activation also induces lymphangiogenesis to facilitate cell migration and enhance antigen, cytokine, and fluid clearance from the affected site. • As a key immune organ, the lymphatics represent a promising target for future lymphatic-directed immunomodulatory therapies and a crucial area of interest for future research into human disease.

This chapter discusses the active roles of the lymphatics in an immune response. Active immune regulation by the peripheral lymphatics is essentially defined as the ability of the lymphatics to dynamically and specifically respond to immune mediators and subsequently modulate the function of peripheral immune cell populations in a stimulus-specific manner. Although these active immune roles remain poorly understood, they are critically important to the understanding of immune functioning as a whole. The peripheral lymphatic system plays several diverse roles in the human body, including the regulation of fluid balance and fatty acid transport from the gastrointestinal tract. Peripheral lymphatic dysfunction can lead to lymphedema, which is generally considered a circulatory condition characterized by abnormal fluid distribution (initially) and fat deposition (later stages). However, evidence of concurrent immune dysfunction in lymphedema suggests that lymphedema could also be classified as a functional immune disorder. The multiple immune deficits in lymphedema 163

K23346_Neligan_11_Immune Reg_r3_dc_0163-0174.indd 163

5/26/2015 8:56:41 AM

164

Part II  Anatomy, Physiology, and Lymphangiogenesis

include excessive fibrosis and local inflammation, poor wound healing, increased susceptibility to infections, and increased risk for the development of malignancy in the affected area.1,2 The appearance of immune dysfunction resulting from lymphatic dysfunction in lymphedema strongly supports a relationship between the lymphatics and the rest of the immune system. This relationship is reciprocal rather than linear, because lymphatic dysfunction is also observed as a result of immune dysfunction in primary immune conditions. For example, chronic inflammation in patients with chronic obstructive pulmonary disease is characterized by multiple immune defects, including abnormalities in the phenotype and density of lymphatic vessels.3 Moreover, altered patterns of lymphangiogenesis (the formation of new lymphatic vessels) have been identified in most human inflammatory conditions, including psoriasis and chronic airway inflammation.4 Finally, peripheral lymphatic vessels also play key roles in transplant rejection and tumor metastases.5 Collectively, these observations highlight the complex and important interconnection between the lymphatic and immune systems. However, despite the identification of peripheral lymphatic involvement at a gross level in many diseases, the specific pathways and mechanisms underlying lymphatic-mediated regulation of immunity remain relatively poorly understood. The primary immune role for the lymphatics is the regulation of cell migration, which is mediated through both the expression of molecules involved in cellular trafficking and the formation of new lymphatic pathways by means of lymphangiogenesis. However, very few studies have directly investigated the immune role of the lymphatics in human disease, and thus the mechanisms of immune regulation remain poorly understood. In addition to this limited understanding, the lymphatics are also still often perceived as passive conduits for fluid, fatty acids, and immune cells. This perception inherently reduces the importance of the lymphatics as an immune organ, and this at least partially explains the omission of the lymphatics in most studies investigating immune function.

Lymphatic Activation by the Immune Microenvironment Compared with a passive component, the essential characteristic of an active component of the immune response is the ability to respond to diverse stimuli and change effector function in response to these diverse stimuli. For example, different activation signals induce different activation programs in macrophages and T cells, such as the classic M1/M2 macrophage responses or Th1/Th2 T-cell responses. This differential integration of signals allows functional specialization and active immune regulation. In contrast to their perceived role as a passive conduit for immune cells, the lymphatics can respond to a wide range of pathogenic stimuli (the so-called primary activation signals) and immune molecules (the so-called secondary activation signals), all of which induce different functional outcomes in immune cell activation/migration and lymphangiogenesis (Fig. 11-1).

Primary Activation Signals Lymphatic endothelial cells express functional toll-like receptors (TLRs) and thus can respond to pathogenic stimuli, including lipopolysaccharide (via TLR4) or lipoteichoic acid (via TLR2).6,7 As with other immune cell populations, stimulation via TLR4 (or any of the pathogen recogni-

K23346_Neligan_11_Immune Reg_r3_dc_0163-0174.indd 164

5/26/2015 8:56:41 AM

Chapter 11  Immune Regulation by the Peripheral Lymphatics

Lymphatic activation with pathogens and immune molecules

165

Induces mediators of immune function and cell migration

Promotes/inhibits lymphangiogenesis



Lymphatic endothelial cell Antigen presenting cell Immune molecules

Pathogenic signals Adhesion molecules Cytokines/chemokines/interleukins

FIG. 11-1  Mechanisms of active immune regulation by the lymphatics. The peripheral lymphatics can respond directly to pathogenic signals and secondary immune mediators produced by pathogen-activated antigen-presenting cells and other immune cell populations. This induces an activation-specific effector program, which involves the regulation of lymphangiogenesis, the expression of cytokines, chemokines, and adhesion molecules for the regulation of cell migration, and interleukins for the regulation of the local immune microenvironment.

tion receptors) induces the expression of a range of inflammatory mediators involved in effector function.6,7 In vitro stimulation of lymphatic endothelial cells with lipopolysaccharide induced the expression of a large number of molecules, including interleukin 6 (IL-6), IL-8, chemokine (CC motif) ligand 21 (CCL21), vascular cell adhesion molecule 1, and intercellular adhesion molecule 1 (ICAM-1).8 Although CCL21, vascular cell adhesion molecule 1, and ICAM-1 are related to the well-appreciated role of the lymphatics in cell migration, IL-6 and IL-8 are classic proinflammatory mediators involved in regulating the function of multiple immune populations,8 suggesting that the lymphatics may also regulate local immune homeostasis. In addition, lymphatic activation does not occur in isolation (as with an in vitro culture model) but instead in a complex, integrated immune milieu. This is perhaps best highlighted in TLRdeficient mouse models, which show concurrent deficiencies in lymphatic architecture/function and macrophage activation/migration. The macrophage deficits are at least partly caused by the reduced ability of the dysfunctional peripheral lymphatics to regulate macrophage recruitment, maturation, and function (and vice versa).9,10 The other crucial observation that highlights the integration of the lymphatics into the normal immune response to pathogens is the ability of lymphatic endothelial cells to uniquely respond to distinct pathogens (as is observed in all immune cell populations). This stimulus-dependent specificity is observed not only in simplified in vitro restimulation assays with various TLR agonists6-8 but also in more physiologic models of inflammation. For example, large differences in the transcriptional expression of key chemokines and integrins were observed in lymphatic endothelial cells isolated from models of oxazolone-induced contact hypersensitivity and complete Freund’s

K23346_Neligan_11_Immune Reg_r3_dc_0163-0174.indd 165

5/26/2015 8:56:42 AM

166

Part II  Anatomy, Physiology, and Lymphangiogenesis

adjuvant-induced inflammation.11 These differences in immune mediator expression by lymphatic endothelial cells contributed to gross differences in inflammation, cell activation/migration, and edema formation between models.11 The large number of differentially expressed genes (.1000) in the inflammation-activated lymphatics also implies a far greater complexity than currently appreciated.11 Thus different pathogenic stimuli induce distinct functional programs in the lymphatics, allowing their active regulation of different immune responses (see Fig. 11-1).

Secondary Activation Signals Although external pathogenic signals may be responsible for the primary initiation of an immune response, a diverse range of interleukins, cytokines, and chemokines (secondary activation signals), produced by activated immune cells, serves to modulate the resulting response. As an integrated component of the immune response, the lymphatics are capable of responding to a wide range of chemokines, interleukins, and other immune mediators with a unique pattern of activation depending on the stimulus.4,12 The effector molecules induced by many of these immune mediators overlap with those induced by pathogenic stimulation,8,13 suggesting that changes in lymphatic phenotype/function in the context of in situ human disease are directed by an inseparable combination of primary and secondary activation signals (see Fig. 11-1). Indeed, it is becoming increasingly clear that activation of all immune populations (now including the lymphatics) in the immune microenvironment is complex and multifaceted and thus likely poorly reproduced in vitro. One of the important consequences of lymphatic activation by secondary immune mediators is the promotion or inhibition of lymphangiogenesis either directly or indirectly through the induction in the expression of lymphangiogenesis-promoting molecules in other immune populations.4,12 In inflammation, lymphangiogenesis increases the density and size of the lymphatic vessels to increase fluid drainage, cell migration, and antigen clearance from the affected site and is intrinsically linked to the resolution or progression of inflammation.14,15 Activated macrophages, T cells, mast cells, and dendritic cells can also directly produce vascular endothelial growth factors (VEGFs) A, C, and D, the canonical mediators of lymphangiogenesis.4,12 Given these strong links between multiple immune modulators and lymphangiogenesis, it is not surprising that altered patterns of lymphangiogenesis are commonly observed in human disease. Collectively, the ability of the lymphatics to regulate their effector function in response to secondary immune mediators emphasizes the lymphatics as an integrated component of the immune response (see Fig. 11-1).

Clinical Significance of Lymphatic Activation Although the ability of the peripheral lymphatics to respond to primary and secondary activation signals per se suggests an important integrated role for the lymphatics in the immune response, several more clinically relevant studies have highlighted a direct link between lymphatic function/ dysfunction and human disease pathogenesis. Lymphatic filariasis (also known as elephantiasis) is a tropical disease caused by the filarial parasites Wuchereria bancrofti (90% of cases), Brugia malayi, and Brugia timori.16 The direct and specific colonization of lymphatic vessels with the filarial parasites induces a clinical disease, which

K23346_Neligan_11_Immune Reg_r3_dc_0163-0174.indd 166

5/26/2015 8:56:42 AM

Chapter 11  Immune Regulation by the Peripheral Lymphatics

167

is remarkably similar to primary/secondary lymphedema, characterized by edema, altered lymphatic vessel distribution/phenotype, increased local fibrosis, and altered immune homeostasis in the affected area16 (see Chapter 15). Direct stimulation of lymphatic endothelial cells with B. malayi has been found to induce the differential expression of several key immunologic molecules related to lymphangiogenesis and immune cell migration and activation.16 This provides a strong link between altered lymphatic activation and the pathogenic characteristics of clinical disease, including the general immune deficits. Similar dysregulation of key immunologic molecules was also observed in lymphatic endothelial cells isolated from patients with lymphedema.17 As with lymphatic filariasis, this result further suggests that intrinsic defects in lymphatic activation are at least partially responsible for the immune dysfunction in patients with lymphedema and lymphatic disease. Thus lymphatic expression of key immune mediators in response to activation signals from the immune microenvironment appears essentially linked to overall immune function. In addition to the direct response to pathogenic signals (such as to filarial parasites), the responsiveness of the lymphatics to secondary immune mediators also appears critically related to lymphatic involvement in the pathogenesis of some human diseases. For example, transforming growth factor beta (TFG-beta) and IL-27 are primarily produced by antigen-producing cells (macrophages and dendritic cells) and have broad immunomodulatory properties, including a net inhibitory effect on lymphatic function and vessel growth.18,19 Altered wound healing is one of the most clinically relevant sequelae of lymphedema in disease morbidity.1 TGF-beta is commonly increased in altered wound healing and has a negative effect on lymphatic regeneration. Because the lymphatics play an important role in the wound healing process, the pathogenesis of altered wound healing in lymphedema may be partially dependent on the effects of altered TGF-beta signaling to the lymphatics, separate from the initial lymphatic insult.18 Similarly, IL-27 has an inhibitory effect on lymphangiogenesis, and circulating levels of IL-27 are commonly observed in many inflammatory disorders.19 Given that altered lymphatic vessel density is also a common characteristic of inflammatory disorders, altered IL-27 signaling to the lymphatics may be one mechanism underlying the lymphatic dysfunction in inflammatory conditions such as psoriasis.19 Thus the altered responses of the peripheral lymphatics to stimulation in human disease appear to be related to disease progression and immune-dependent pathogenesis.

Lymphatic Regulation of the Immune Response The definition of the lymphatics as an active component depends not only on their ability to respond to diverse stimuli but also on their ability to actively modulate the function of other immune cells (Box 11-1). As a conduit system connecting the peripheral to the lymphoid organs, the primary immune role of the lymphatics is the regulation of cell migration and migratory cell activation/function. However, the lymphatics also appear to play a role in regulating local immune responses, separate from immune cell migration. Moreover, the lymphatics have an important role in draining lo-

K23346_Neligan_11_Immune Reg_r3_dc_0163-0174.indd 167

5/26/2015 8:56:42 AM

168

Part II  Anatomy, Physiology, and Lymphangiogenesis

BOX 11-1  Immune Processes With Peripheral Lymphatic Involvement • Drainage of local immune mediators from the tissue site • Immune cell activation/maturation • Immune cell migration • Immune tolerance induction • Regulation of local immune responses • Removal of cytokines and chemokines from circulation • Self- (tolerance) and foreign (immunity) antigen transport • Tissue fibrosis • Transplant rejection

cal immune molecules from the tissue site to maintain homeostasis of the local immune site and prevent damage from the accumulation of these immune mediators.20 The lymphatics are also involved in the direct removal of cytokines and chemokines from circulation by the expression of scavenger receptors such as D6 and the transport of antigens to lymphoid organs for immune tolerance (to peripheral self-antigens) or immune activation (to soluble foreign antigens).20 These additional roles for the lymphatics are critically linked to lymphangiogenesis and its regulation by the secondary immune mediators present in most immune responses as discussed previously.

Regulation of Cellular Migration and Activation The migration of dendritic cells from an infected tissue site to the draining lymph node is the essential first step in the initiation of peripheral adaptive immune responses. Despite the perception of the lymphatics as an inert conduit for cell migration, the lymphatics actively regulate dendritic cell migration and activation and thus can be considered an essential component of the adaptive immune system. Dendritic cell migration in the steady state and during inflammation involves different mechanisms that have largely been attributed to activation-induced expression of different chemokine receptors and integrins on dendritic cells.21 However, there is a concurrent, stimulus-specific upregulation of the chemokine and integrin partners on the peripheral lymphatics during inflammation.4,8,12,13 In addition, activation induces functional and spatial reorganization of peripheral lymphatic vessels (for example, the formation of ICAM-1–enriched microvilli structures) to actively promote cell migration.22 Thus lymphatic activation and functional reorganization are critical steps in the active regulation of dendritic cell immunity (Fig. 11-2). In addition to regulating migration into the lymph nodes, the lymphatics also contribute to migratory dendritic cell maturation/function (see Fig. 11-2). Signaling through chemokine receptors (for example, CCL21 through CCR7) and integrins (for example, ICAM-1 through lymphocyte function-associated antigen 1 [LFA-1]) induces both migration and maturation in dendritic cells and other immune cell subsets.23,24 Mice deficient for the agonists CCR7 through CCL21 show abnormal dendritic cell maturation, including altered proliferation, endocytosis, differentiation, and survival, which is separate from the defects in dendritic cell migration.23 In another example, steady-state dendritic cells receiving signals through lymphatic-expressed ICAM-1 show reduced CD86 expression, altered maturation, and a consequent reduction in their ability to activate T

K23346_Neligan_11_Immune Reg_r3_dc_0163-0174.indd 168

5/26/2015 8:56:42 AM

Chapter 11  Immune Regulation by the Peripheral Lymphatics

Lymphatic endothelial cell Dendritic cell Immune cell

169

Immune molecules Pathogenic signals Adhesion molecules Cytokines/chemokines/interleukins

FIG. 11-2  An integrated model of immune cell activation. Concurrent activation of all local immune cell populations by pathogenic stimuli and pathogen-induced immune mediators allows coordinated immune cell activation and a fully functional immune response. Thus the peripheral lymphatics are one component of a tightly integrated immune system in which multiple immune populations coordinate their functional responses to pathogens and other immune insults, such as cancer and trauma.

cells.24,25 These results suggest that the interaction between the lymphatic endothelial cells and dendritic cells is critical for dendritic cell maturation and function, including downstream regulation of T-cell tolerance. Given that T cells also traffic through the lymphatics at various stages of the immune response, it is logical to suggest that the lymphatics play a similar active role in dendritic cell migration. Indeed, various T-cell subsets also express CCR7, and thus their maturation is also influenced by lymphatic expression of CCL21.26 Similarly, lymphatic-expressed molecules such as CCL21 can also induce local macrophage recruitment and maturation (without migration through the lymphatics).9 Finally, the lymphatic regulation of local immune function and immune cell maturation is not restricted to secondary cell activation in response to molecules primarily involved in migration. The lymphatics can also express a wide range of polyfunctional proinflammatory and antiinflammatory molecules, including IL-6/IL-8, which regulate neutrophil, macrophage, and dendritic cell function,8 and IL-7, which is involved in T-cell homeostasis.27 Although our understanding of the lymphatic-dependent regulation of immune function remains limited, these observations support a multifunctional, active role for the lymphatics in regulating the immune response both in cell migration and local immune function (see Fig. 11-2).

Clinical Significance of Lymphatic Regulation of Immune Responses Given the lack of definitive information about the active roles of the lymphatics in regulating immune responses, it remains difficult to determine the clinical significance of lymphatic dysfunction in disease. However, given the increasingly appreciable role for the lymphatics in normal dendritic

K23346_Neligan_11_Immune Reg_r3_dc_0163-0174.indd 169

5/26/2015 8:56:42 AM

170

Part II  Anatomy, Physiology, and Lymphangiogenesis

cell, macrophage, and T-cell biology, it is extremely likely that the immune dysfunction observed in diseases showing altered lymphatic function is at least partially attributable to lymphatic intrinsic deficits in immune modulation. This also implies that lymphatic-targeted therapeutic approaches are a potential tool to address immune dysfunction in clinical practice. Several possible therapies have been proposed based on the ability of the lymphatic system to regulate immune function. One such approach is to alter T-cell responses by indirectly targeting dendritic cell maturation/migration through lymphatic promotion or inhibition.12,21 This approach might be useful in dendritic cell–delivered vaccinations in which enhanced migration/maturation is desired or in transplantation in which inhibiting migration might prevent graft rejection.12,21 Targeting the lymphatics in inflammatory disorders is another promising therapy for modulating other aspects of the immune regulatory functions of the lymphatics. Although these and other intervention strategies have, for the most part, not been introduced into clinical practice, they remain an area of increasing promise. As our understanding of lymphatic-based regulation of the immune response increases, the number of therapeutic targets and therapeutic options should also increase.

Linking Lymphatic Damage to the Immune Pathogenesis of Lymphedema Lymphedema is generally initiated by local lymphatic damage after radiation or surgery but results in several global immune deficits, including defects in immune surveillance (susceptibility to infection and malignancy) and local immune homeostasis (abnormal fibrosis, inflammation, and wound healing).1,2 Although these immune deficits are commonly observed in patients with lymphedema, the mechanisms of disease progression after an initial lymphatic insult are poorly understood. However, the active immune roles of the peripheral lymphatics may provide an insight into the pathogenesis of lymphedema. The primary result of lymphatic vessel damage after interventions such as surgery or radiation therapy is a gross reduction in drainage function.1,2 Given the responsiveness of the peripheral lymphatics to local immune mediators, local accumulation of fluid and immune mediators is likely to contribute to the gross transcriptional differences observed in lymphatic endothelial cells isolated from patients with lymphedema.17 Because the peripheral lymphatics appear to be critically involved in the regulation of local immune cell migration, maturation, and function, the altered lymphatic phenotype may contribute to the observed defects in immune surveillance (resulting from the dysfunctional lymphatic regulation of dendritic cell function and/or reduced antigen transport to the lymph nodes) and immune homeostasis (resulting from the altered immune mediator production by the lymphatics and/or deficient drainage of molecules from the local site). However, given the tightly integrated nature of the immune system (see Fig. 11-2), direct alterations in the function of other immune populations (resulting from abnormal immune mediator accumulation) also play an essential role in disease progression. Collectively, the reduction in the “passive” drainage function appears to be the initiating factor in the pathogenesis of lymphedema through local dysregulation of immune homeostasis. However, the subsequent alterations in the “active” immune regulatory functions of the lymphatics may also be critical for the progression of lymphedema.

K23346_Neligan_11_Immune Reg_r3_dc_0163-0174.indd 170

5/26/2015 8:56:42 AM

Chapter 11  Immune Regulation by the Peripheral Lymphatics

171

Conclusion The lymphatics are an active and integrated component of the immune response. Their active role in the immune response is initiated by the recognition of pathogenic and immunogenic stimuli by the peripheral lymphatics. These stimuli induce stimulus-specific activation programs in the lymphatics that allow the coordination of the immune functions of multiple additional immune populations. Alterations in these processes in lymphedema resulting from abnormal lymphatic vessel activation may be one contributing factor to the gross immune deficiencies that are characteristic of lymphedema. In general, the identification of the lymphatics as a critical immune organ has major consequences for our interpretation of immunity in health and disease in the following ways: • Although lymphatic dysfunction has been widely identified in a range of human immune conditions, the mechanisms, sequelae, and relative importance of this dysfunction remain poorly understood. • The peripheral lymphatics function as a core, integrated component of the peripheral immune system, and an improved understanding of their role is crucial for a complete appreciation of the immune processes involved in human disease. This necessitates a greater focus on lymphatic function in immunologic studies to clarify these roles, especially because the lymphatics are perhaps the least well-studied component of the immune system. • Lymphatics make up one component of a tightly coordinated immune system involving multiple immune cell populations. • The inherent implication of this observation is that multiple players are involved in any given aspect of an immune response, and scientific studies, where possible, must be designed and interpreted with this in mind. For example, a study of peripheral dendritic cell activation of T cells in inflammation is likely not complete without also considering the influence of lymphatic modulation on the dendritic cell’s function. • Overall, an increased understanding of the roles of the peripheral lymphatics is required to fully appreciate human disease and open up the possibilities for lymphatic-targeted interventions.

C linical P earls • A dysfunctional lymphatic system after surgery or radiation can lead to the development of lymphedema. • Later stages of lymphedema are characterized by an increased susceptibility to infection/malignancy and altered patterns of fibrosis, wound healing, and inflammation. • Dysfunctional lymphatic vessels have also been identified in a wide range of conditions, ranging from psoriasis to cancer, and may play a role in the altered immune homeostasis in these conditions. • A comprehensive clinical understanding of any human immune pathology may require consideration of the role of the lymphatic system. • As such, the lymphatic system may serve as a novel therapeutic target in human immune modulatory medicine.

K23346_Neligan_11_Immune Reg_r3_dc_0163-0174.indd 171

5/26/2015 8:56:42 AM

172

Part II  Anatomy, Physiology, and Lymphangiogenesis

R EFERENCES 1. Rockson SG. The lymphatics and the inflammatory response: lessons learned from human lymphedema. Lymphat Res Biol 11:117-120, 2013. 2. Ruocco V, Schwartz RA, Ruocco E. Lymphedema: an immunologically vulnerable site for development of neoplasms. J Am Acad Dermatol 47:124-127, 2002. 3. Mori M, Andersson CK, Graham GJ, et al. Increased number and altered phenotype of lymphatic vessels in peripheral lung compartments of patients with COPD. Respir Res 14:65, 2013. 4. Aebischer D, Iolyeva M, Halin C. The inflammatory response of lymphatic endothelium. Angiogenesis 17:383-393, 2014. 5. Pegu A, Qin S, Fallert Junecko BA, et al. Human lymphatic endothelial cells express multiple functional TLRs. J Immunol 180:3399-3405, 2008. 6. Sawa Y, Tsuruga E. The expression of E-selectin and chemokines in the cultured human lymphatic endothelium with lipopolysaccharides. J Anat 212:654-663, 2008. 7. Sawa Y, Tsuruga E, Iwasawa K, et al. Leukocyte adhesion molecule and chemokine production through lipoteichoic acid recognition by toll-like receptor 2 in cultured human lymphatic endothelium. Cell Tissue Res 333:237-252, 2008. 8. Sawa Y, Ueki T, Hata M, et al. LPS-induced IL-6, IL-8, VCAM-1, and ICAM-1 expression in human lymphatic endothelium. J Histochem Cytochem 56:97-109, 2008. 9. Kang S, Lee SP, Kim KE, et al. Toll-like receptor 4 in lymphatic endothelial cells contributes to LPSinduced lymphangiogenesis by chemotactic recruitment of macrophages. Blood 113:2605-2613, 2009. 10. Zampell JC, Elhadad S, Avraham T, et al. Toll-like receptor deficiency worsens inflammation and lymphedema after lymphatic injury. Am J Physiol Cell Physiol 302:C709-C719, 2012. 11. Vigl B, Aebischer D, Nitschke M, et al. Tissue inflammation modulates gene expression of lymphatic endothelial cells and dendritic cell migration in a stimulus-dependent manner. Blood 118:205-215, 2011. 12. Dieterich LC, Seidel CD, Detmar M. Lymphatic vessels: new targets for the treatment of inflammatory diseases. Angiogenesis 17:359-371, 2014. 13. Chaitanya GV, Franks SE, Cromer W, et al. Differential cytokine responses in human and mouse lymphatic endothelial cells to cytokines in vitro. Lymphat Res Biol 8:155-164, 2010. 14. Huggenberger R, Siddiqui SS, Brander D, et al. An important role of lymphatic vessel activation in limiting acute inflammation. Blood 117:4667-4678, 2011. 15. Kataru RP, Jung K, Jang C, et al. Critical role of CD11b1 macrophages and VEGF in inflammatory lymphangiogenesis, antigen clearance, and inflammation resolution. Blood 113:5650-5659, 2009. 16. Bennuru S, Nutman TB. Lymphangiogenesis and lymphatic remodeling induced by filarial parasites: implications for pathogenesis. PLoS Pathog 5:e1000688, 2009. 17. Ogunbiyi S, Chinien G, Field D, et al; London Lymphedema Consortium. Molecular characterization of dermal lymphatic endothelial cells from primary lymphedema skin. Lymphat Res Biol 9:19-30, 2011. 18. Clavin NW, Avraham T, Fernandez J, et al. TGF-beta 1 is a negative regulator of lymphatic regeneration during wound repair. Am J Physiol Heart Circ Physiol 295:H2113-H2127, 2008. 19. Nielsen SR, Hammer T, Gibson J, et al. IL-27 inhibits lymphatic endothelial cell proliferation by STAT1regulated gene expression. Microcirculation 20:555-564, 2013. 20. Santambrogio L, Stern LJ. Carrying yourself: self antigen composition of the lymphatic fluid. Lymphat Res Biol 11:149-154, 2013. 21. Teijeira A, Russo E, Halin C. Taking the lymphatic route: dendritic cell migration to draining lymph nodes. Semin Immunopathol 36:261-274, 2014. 22. Teijeira A, Garasa S, Peláez R, et al. Lymphatic endothelium forms integrin-engaging 3D structures during DC transit across inflamed lymphatic vessels. J Invest Dermatol 133:2276-2285, 2013.

K23346_Neligan_11_Immune Reg_r3_dc_0163-0174.indd 172

5/26/2015 8:56:42 AM

Chapter 11  Immune Regulation by the Peripheral Lymphatics

173

23. Marsland BJ, Bättig P, Bauer M, et al. CCL19 and CCL21 induce a potent proinflammatory differentiation program in licensed dendritic cells. Immunity 22:493-505, 2005. 24. Podgrabinska S, Kamalu O, Mayer L, et al. Inflamed lymphatic endothelium suppresses dendritic cell maturation and function via Mac-1/ICAM-1-dependent mechanism. J Immunol 183:1767-1779, 2009. 25. Wilson NS, Young LJ, Kupresanin F, et al. Normal proportion and expression of maturation markers in migratory dendritic cells in the absence of germs or Toll-like receptor signaling. Immunol Cell Biol 86:200-205, 2008. 26. Masopust D, Schenkel JM. The integration of T cell migration, differentiation and function. Nat Rev Immunol 13:309-320, 2013. 27. Miller CN, Hartigan-O’Connor DJ, Lee MS, et al. IL-7 production in murine lymphatic endothelial cells and induction in the setting of peripheral lymphopenia. Int Immunol 25:471-483, 2013.

K23346_Neligan_11_Immune Reg_r3_dc_0163-0174.indd 173

5/26/2015 8:56:42 AM

K23346_Neligan_11_Immune Reg_r3_dc_0163-0174.indd 174

5/26/2015 8:56:42 AM

Part III

Pathophysiology and Clinical Presentation

K23346_Neligan_12_Pathophysiology_r3_0175-0188.indd 175

5/26/2015 9:02:38 AM

K23346_Neligan_12_Pathophysiology_r3_0175-0188.indd 176

5/26/2015 9:02:38 AM

C hapter 12 Pathophysiology of Primary Lymphedema Byung-Boong Lee, James Laredo

K ey P oints • Primary lymphedema usually manifests as a truncular lymphatic malformation, the origin of which is a structural birth defect. • Although most primary lymphedemas are independent truncular lymphatic malformations, they can coexist with extratruncular lesions. • Occasionally, primary lymphedema and truncular lymphatic malformations develop along with congenital vascular malformations to form a hemolymphatic malformation.

Pat

• The defective development of the lymphatic system can occur at any stage of lymphangiogenesis, even as early as the eighth week of gestation. • In primary lymphedema the cause of reduced lymph transport is either an intrinsic defect or malfunction of the lymph conducting elements that result from a genetic abnormality in lymphatic function or anatomy.

Primary lymphedema mostly presents as the clinical manifestation of a truncular lymphatic malformation, with defective structural development of the lymphatic system observed as a birth defect.1-4 Most primary lymphedemas exist alone as an independent truncular lymphatic malformation lesion. However, because of the pathogenesis of congenital vascular malformations (CVMs), some of the primary lymphedemas and truncular lymphatic malformations exist with another form of lymphatic malformation, the extratruncular lesions, also known as cystic, cavernous, or capillary lymphangiomas.5-8 In addition, lymphatic malformations exist with other types of CVMs as part of complex birth defects affecting the entire circulation, including the arteries, veins, lymphatics, and capillary system.9-12 Therefore primary lymphedemas and truncular lymphatic malformations infrequently develop together with other CVMs to form a hemolymphatic malformation,13-16 consisting of a venous malformation,17-20 an arteriovenous malformation,21-24 and/or a capillary malformation.25,26 177

K23346_Neligan_12_Pathophysiology_r3_0175-0188.indd 177

5/26/2015 9:02:38 AM

178

Part III  Pathophysiology and Clinical Presentation

For example, the lymphatic malformation coexists with venous and lymphatic malformations as the vascular malformation component of Klippel-Trenaunay syndrome.27,28 When they are further combined with an arteriovenous malformation, it is Parkes Weber syndrome. 29,30 The defective development of the lymphatic system can occur throughout any stage of lymphangiogenesis, resulting in various malformations of the lymphatic system, all of which have a basic outcome—lymphatic dysfunction.31-34 As early as the eighth week of gestation, the developing lymphatic sacs, which consist of two jugular and two iliac sacs, one of which is positioned at the base of the root of the mesentery and the other dorsal to the abdominal aorta/cisterna chyli, can show defective maturation. This results in sequestration of primitive lymphatic tissue; it remains as isolated clusters of amorphous lymphatic tissues (for example, lymphangioma).35,36 When these primitive lymphatic tissues do not develop normally and communicate with the remainder of the lymphatic networks and are consequently sequestered in various regions, they continue to dilate and become macrocystic and microcystic lesions, such as a cystic hygroma.37-40 This malformed lymphatic tissue has the unique characteristic of the mesenchymal cell when stimulated and appears as a residual embryonic tissue remnant originating from its early stage of lymphangiogenesis. These premature lymphatic tissues are classified as extratruncular lesions of the lymphatic malformation and are the outcome of defective development after the developmental arrest in the early stage of lymphangiogenesis.41-44 Extratruncular lymphatic malformation lesions, often called lymphangioma, continue to grow after birth through the unique characteristics of mesenchymal cells and lymphangioblasts (by way of comparison, truncular lymphatic malformation) whenever conditions are suitable. Some examples include menarche, pregnancy, female hormones, trauma, and surgery. After the ninth week of gestation, when disturbances in the lymph nodes and vessel formation processes associated with the main lymphatic trunk formation occur, various and often more significant structural variants of the lymph transporting system take place, including those of the thoracic duct.45,46 Such defective development during the later stage of lymphangiogenesis before the completion of fully mature normal lymphatic trunk formation is also grouped separately as truncular lymphatic malformations. These truncular lesions no longer have the ability to grow like extratruncular lesions (as previously described), but they can have a much more serious impact overall with respect to lymph transport function because of their direct structural impact on the lymph vessels.47,48 Therefore primary lymphedema mainly presents as a clinical manifestation of these defective formations of the lymphatic trunk, vessels, and nodes from the late stage of lymphangiogenesis. We see a range of defects, including lymphatic trunk hypoplasia, aplasia, numeric hyperplasia, or dilation (lymphangiectasia) with valvular incompetence. Regarding the lymph nodes, we often see selective lymph node dysplasia alone, which is also indicative of primary lymphedema, and is best known as lymphnododysplasia, although it is generally involved with lymphangiodysplasia as indicated previously.49,50

K23346_Neligan_12_Pathophysiology_r3_0175-0188.indd 178

5/26/2015 9:02:38 AM

Chapter 12  Pathophysiology of Primary Lymphedema

179

To add complexity, not all primary lymphedemas are associated with such anatomic defects, or if present, they seemingly have little effect. Instead, some show only defective function, such as Milroy disease.51-54 Indeed, some primary lymphedemas have very little structural derangement and are limited to a functional defect that appears molecular in origin. For these reasons, some investigators believe that all lymphedemas caused by lymphatic malformations are genetically derived, and they define lymphedema as an abnormality of lymph drainage in which the predominant effect is on the tissue territory(ies) drained. Therefore a malformation may not necessarily have attributes that can be imaged, but may ultimately require detection or definition in molecular or other functional terms.3,4,55,56 Thus the cause of decreased lymphatic transport in primary lymphedema is either an intrinsic defect 5,8,57-60 or a malfunction of the lymph-conducting elements61-64 resulting from a genetically determined abnormality of lymphatic anatomy or function. When primary chronic lymphedema represents the clinical expression of heritable abnormal structural development, it clinically manifests as a macroscopic structural abnormality, which is defined as a truncular lymphatic malformation.8,65 Furthermore, primary lymphedema includes all the lymphedemas caused by the mutation of any of the genes involved in the development of the lymphatic system. Currently, representative genetic mutations identified in a familial distribution of primary lymphedema are FLT4,52,57,66 FoxC,54,57 and GJC2.67,68 The major clinical sign of all these forms of genetically determined lymphedema is lymphedema as the primary phenotype. For these families, an autosomal dominant pattern of inheritance has been reported. Various genes strongly associated with this pattern of inheritance have been demonstrated, with variable expression and variable age at onset.56,63,66 Milroy disease is one example of a familial lymphedema caused by an autosomal dominant, single gene disorder inherited as a germ line mutation at the locus 5q35.3. The gene mutated is FLT4, which encodes for the vascular endothelial growth factor receptor 3 (VEGFR-3).56,66,69 However, because of a somatic mutation, regionally limited genetic disorders will allow some tissue (for example, skin) to be unaffected, whereas the adjacent tissue (skin) carries the mutation (for example, mosaicism). Nevertheless, the hereditary type of primary lymphedema is quite rare, whereas the sporadic type represents the majority, although both have a genetic basis. What is important, however, is to acknowledge that all the malformations by definition are present at birth, although there is some concern regarding the current definition of primary lymphedema, because postnatal obliterations of lymph collectors and lymph nodes can mimic congenital and prenatal pathologies.

Overcoming the Problem Various cytokines70,71 are involved in the stimulation of lymphangiogenesis. For instance, vascular endothelial growth factor (VEGF)-C and -D are known to activate VEGFR-3 expressed on lymphatic endothelial cells.72 VEGF-C–deficient mice fail to develop a functional lymphatic system,73 attesting to its importance.

K23346_Neligan_12_Pathophysiology_r3_0175-0188.indd 179

5/26/2015 9:02:38 AM

180

Part III  Pathophysiology and Clinical Presentation

Angiopoietin 1 also promotes lymphatic vessel formation through Tie2.74 Also, gene transfer of VEGF-C through its promotion of lymphangiogenesis reduces lymphedema, at least in an animal model.75 For these reasons, patients with lymphatic malformations with such risks for several congenital lymphatic disorders, including Down syndrome, Turner syndrome, and Noonan syndrome in particular, should receive genetic counseling for cytogenetic analysis for chromosomal aneuploidy before they consider becoming parents, because aneuploidic conditions can recur in subsequent pregnancies.76-79 As the previously mentioned case demonstrates, an accurate definition of the normal anatomophysiology of the lymphatic system is essential for a proper understanding of the pathophysiology of primary lymphedema and perhaps for better management of the condition.

Normal Role of the Lymphatics and Impact of Defects on the Cells, Tissue, and Body The circulatory function is the principal role of the lymphatic system, which maintains the drainage and transport of interstitial fluid to the main blood circulation. However, in addition to such an essential role for interstitial fluid homeostasis, the lymphatic system has multiple roles. It plays a crucial role for the immune systems, providing the immune traffic route to transport white blood cells and antigen-presenting cells to the lymphoid organs.80 It also has another unique function for the lipid absorption from the gastrointestinal tract. Absorption of fat from the intestine occurs through the lymphatic system, and subsequent transportation of the chyle to the liver depends on this system.81-83 Such crucial functions are directly affected by a defective development no matter what the reason. Many if not most of the structural malformations previously described lead to functional issues. The lymphatics are found throughout the body except in the central nervous system, and lymphatic vasculature and lymphoid tissue are more prevalent in tissues that are in close contact with the external environment (for example, the gastrointestinal tract and lungs).84 Such distribution seems to reflect the unique role of the lymphatics against infectious agents and foreign materials. Thus the location of the malformations may have an impact on any or all tissues. In the extremities the lymphatic system consists of superficial and deeper systems. A superficial system collects lymph from the epifascial tissue, such as skin and subcutaneous tissue, whereas a deeper system drains subfascial tissue, such as muscle, bone, and other deep structures like blood vessels. The two systems are mutually interdependent to provide the compensatory function, especially when one system fails because of a structural malformation or functional failure associated with it to maintain normal function and drainage. For example, the deep lymphatic system participates in lymph transport from the skin during lymphatic obstruction.85 The failure of adequate lymph transport promotes lymphedema and likely contributes to the pathologic presentation of a wide variety of lymphatic vascular diseases. In a normal state, the

K23346_Neligan_12_Pathophysiology_r3_0175-0188.indd 180

5/26/2015 9:02:38 AM

Chapter 12  Pathophysiology of Primary Lymphedema

181

extravasation of fluids and proteins from blood vessels is adequately handled by lymphatic drainage and return into the bloodstream. However, if microvascular filtration in blood capillaries and venules exceeds the capacity of lymphatic drainage for long periods, an edema develops because of the accumulation of interstitial fluid in the interstitium (for example, advanced chronic venous disease).3 Lymph flow is established by the autonomous rhythmic contractions of the lymphangions, which propel the lymph.86-88 When interstitial fluid enters the initial lymphatics to flow into the lymphangions, the lymphatic wall is stretched by inflowing interstitial fluid. It stimulates the lymphatic wall muscles to evoke the contractions to generate the flow in a peristaltic fashion. The frequency of rhythmic contraction of the lymphatics depends on the volume of the interstitial fluid entering.88,89 Fluid transport into the initial lymphatics occurs against a pressure gradient. Such a phenomenon is explained based on the condition when episodic increases in interstitial fluid pressure by tissue movement combine with suction forces generated through the contraction of the collecting lymphatics.90 Under normal conditions, muscular activity, respiratory movements, passive movements, and arterial pulsation have little if no effect on lymph flow when the lymphangion-based peristaltic movement is sufficient for lymph transport and flow.87-89 Therefore the lymphatics of the limb are generally empty, with only a few microliters of lymph in some lymphangions. There is no hydrostatic pressure in the lymphatics of a normal leg in the upright position.88,89 The normal lymph transport mechanism is based on the autoregulated peristaltic flow led by the lymphangions; however, in lymphedema this mechanism fails. In addition, the unique lymphodynamics that would be applicable to normal conditions are no longer valid, and new fluid dynamics to compensate for the lymphedematous condition become identical to the venodynamics. Thus muscular contraction of the foot and calf may increase lymph pressure, and patent lymphatics are filled with lymph. Compression of the muscles could create a pressure gradient between the distal and proximal lymphatics.88,89 In normal limbs, the lymph flow occurs only during spontaneous contractions of lymphangions.89 However, in lymphedematous limbs, most of the lymph collectors are partially or totally obliterated as a consequence of the destruction of lymph vessel musculature and valves, and only some spontaneous flow may remain in patent vessel segments at different levels of the limb.91-93

Lymphostasis Various types of congenital abnormalities of lymphatic vessels and lymph nodes often lead to lymphatic hypertension, valvular insufficiency, and lymphostasis. An accumulation of interstitial and lymphatic fluid within the skin and subcutaneous tissue stimulates fibroblasts, keratinocytes, and adipocytes. Such a condition eventuates in the deposition of collagen and glycosaminoglycans within the skin and subcutaneous tissue to result in the destruction of elastic fibers and skin hypertrophy.94 The tissues deprived of proper lymph drainage become the site of a continuous inflammatory process.

K23346_Neligan_12_Pathophysiology_r3_0175-0188.indd 181

5/26/2015 9:02:38 AM

182

Part III  Pathophysiology and Clinical Presentation

Lymphedema fluids accumulating in the skin and subcutaneous tissues contain cytokines, chemokines, activated immune cells, and most important, microorganisms. Lymph cytokines are lowmolecular-weight proteins, and thus they easily penetrate the capillary wall. However, they are also produced locally by keratinocytes, fibroblasts, dermal macrophages, dendritic cells, and lymphocytes in the skin and subcutaneous tissue. The local production increases the lymph cytokine levels to those above plasma (for example, lymphatic endothelial cell–produced VEGF- C).95,96 Under normal conditions, circulating immune cells are able to eliminate the microorganisms penetrating the epidermis, but in lymph stasis, for instance, they are unable to remove the microorganisms. Thus the microorganisms still proliferate to cause histologic changes, such as infiltrates and the formation of fibrous tissue, known as the clinical condition of dermatolymphangioadenitis.96 In lymphedema, the pathologic changes in the lymphatics resulting from infection can include a plethora of damage to the endothelial and muscular cells, which may subsequently lead to an obliteration of the lumen by the fibroblasts—the price the lymphatic system pays for its own function. The pathologic events in the skin and the reaction of the regional lymphatic system to them are the outcomes of the destructive effect of the inflammatory process to the healing of parenchymatous tissues caused by the system devoted to the elimination of microbes and clearance of damaged cells.97-99 There are all the other factors and influences, which in some ways may, despite the well-intentioned process, continue to worsen or compromise its function. The breathing space or reserve capacity of the lymphatics that have a primary deficiency or defect or dysfunction is less, and thus a patient with these circumstances who has an underlying primary lymphatic issue must be even more careful of these other issues, which may further compromise the lymphatic system and the range of critical roles it performs.

Conclusion The typical clinical presentation of primary lymphedema is a truncular lymphatic malformation with defective structural development of the lymphatic system that occurs during the later stage of lymphangiogenesis before fully mature normal lymphatic trunk formation is complete. Thus primary lymphedema usually results from a defect in the formation of the lymphatic trunk, vessels, and nodes and occurs in a wide range of defects, including lymphatic trunk hypoplasia, aplasia, numeric hyperplasia, or dilation (lymphangiectasia) with valvular incompetence. However, not all primary lymphedemas are associated with these anatomic defects. Some have only defective or limited function with very little structural derangement, such as Milroy disease. Therefore the cause of decreased lymphatic transport in primary lymphedema is either an intrinsic defect or a malfunction of the lymph-conducting elements resulting from a genetically determined abnormality of lymphatic anatomy or function. Most primary lymphedema and truncular lymphatic malformations exist alone as an independent lesion, but occasionally they are found with another form of lymphatic malformation—extratruncular lesions—also known as cystic, cavernous, or capillary lymphangiomas.

K23346_Neligan_12_Pathophysiology_r3_0175-0188.indd 182

5/26/2015 9:02:39 AM

Chapter 12  Pathophysiology of Primary Lymphedema

183

Extratruncular lesions are residual embryonic tissue remnants that are the outcome of defective development from its early stage of lymphangiogenesis. Thus they have the unique characteristic of the mesenchymal cell; they grow steadily through the rest of life by various stimulations (by way of comparison, truncular lesions). In addition, lymphatic malformations exist with other types of CVMs as a part of complex birth defects that affect the entire circulation involving the arteries, veins, lymphatics, and capillary system.

C linical P earls • Most primary lymphedemas are truncular malformations. However, they can coexist with extratruncular lesions, also called cystic, cavernous, or capillary lymphangiomas. • Most primary lymphedemas are a defect of the lymphatic trunk, vessels, and nodes that occurs during late stage lymphangiogenesis. However, some primary lymphedemas have a molecular or genetic origin. • Patients at risk of severe congenital lymphatic disorders, such as Down syndrome, should undergo genetic counseling if they are considering becoming parents. • An inadequate lymph transport mechanism promotes lymphedema, contributes to various lymphatic vascular disorders, and causes edema. • Improper lymph drainage results in a continuous inflammatory process of tissue, including infection.

R EFERENCES 1. Lee BB, Villavicencio JL. Primary lymphedema and lymphatic malformation: are they the two sides of the same coin? Eur J Vasc Endovasc Surg 39:646-653, 2010. 2. Lee BB, Andrade M, Bergan J, et al; International Union of Phlebology. Diagnosis and treatment of primary lymphedema. Consensus Document of the International Union of Phlebology (IUP)-2009. Int Angiol 29:454-470, 2010. 3. Lee BB, Andrade M, Antignani PL, et al; International Union of Phlebology. Diagnosis and treatment of primary lymphedema. Consensus Document of the International Union of Phlebology (IUP)-2013. Int Angiol 32:541-574, 2013. 4. Lee BB, Antignani PL, Baroncelli TA, et al. IUA-ISVI consensus for diagnosis guideline of chronic lymphedema of the limbs. Int Angiol. 2014 Mar 19. [Epub ahead of print] 5. Lee BB. Lymphedema-angiodysplasia syndrome: a prodigal form of lymphatic malformation (LM). Phlebolymphology 47:324-332, 2005. 6. Lee BB, Laredo J, Lee TS, et al. Terminology and classification of congenital vascular malformations. Phlebology 22:249-252, 2007. 7. Lee BB, Laredo J, Lee SJ, et al. Congenital vascular malformations: general diagnostic principles. Phlebology 22:253-257, 2007. 8. Lee BB, Kim YW, Seo JM, et al. Current concepts in lymphatic malformation (LM). Vasc Endovasc Surg 39:67-81, 2005.

K23346_Neligan_12_Pathophysiology_r3_0175-0188.indd 183

5/26/2015 9:02:39 AM

184

Part III  Pathophysiology and Clinical Presentation

9. Lee BB. Congenital venous malformation: changing concept on the current diagnosis and management. Asian J Surg 22:152-154, 1999. 10. Lee BB, Bergan JJ. Advanced management of congenital vascular malformations: a multidisciplinary approach. J Cardiovasc Surg 10:523-533, 2002. 11. Lee BB. Critical issues on the management of congenital vascular malformation. Ann Vasc Surg 18:380392, 2004. 12. Lee BB, Mattassi R, Loose D, et al. Consensus on controversial issues in contemporary diagnosis and management of congenital vascular malformation—Seoul communication. Int J Angiol 13:182-192, 2004. 13. Lee BB, Laredo J. Classification: venous-lymphatic vascular malformation. In Allegra C, Antignani PL, Kalodiki, eds. News in Phlebology. Turin, Italy: Edizioni Minerva Medica, 2013. 14. Lee BB, Bergan J, Gloviczki P, et al; International Union of Phlebology. Diagnosis and treatment of venous malformations. Consensus Document of the International Union of Phlebology (IUP)-2009. Int Angiol 28:434-451, 2009. 15. Lee BB, Baumgartner I, Berlien P, et al. Diagnosis and Treatment of Venous Malformations Consensus Document of the International Union of Phlebology (IUP): updated 2013. Int Angiol. 2014 Feb 25. [Epub ahead of print] 16. Lee BB, Antignani PL, Baraldini V, et al. ISVI-IUA consensus document. Diagnostic guidelines on vascular anomalies: vascular malformations and hemangiomas. Int Angiol. 2014 Oct 6. [Epub ahead of print] 17. Lee BB. Venous malformation and haemangioma: differential diagnosis, diagnosis, natural history and consequences. Phlebology 28 Suppl 1:176-187, 2013. 18. Lee BB, Laredo J. Venous malformation: treatment needs a bird’s eye view. Phlebology 28:62-63, 2013. 19. Lee BB, Baumgartner I. Contemporary diagnosis of venous malformation. J Vasc Diagn 1:25-34, 2013. 20. Lee BB. Current concept of venous malformation (VM). Phlebolymphology 43:197-203, 2003. 21. Lee BB, Lardeo J, Neville R. Arterio-venous malformation: how much do we know? Phlebology 24:193200, 2009. 22. Lee BB, Baumgartner I, Berlien HP, et al; International Union of Angiology. Consensus Document of the International Union of Angiology (IUA)-2013. Current concept on the management of arteriovenous management. Int Angiol 32:9-36, 2013. 23. Lee BB, Mattassi R, Kim BT, et al. Contemporary diagnosis and management of venous and AV shunting malformation by whole body blood pool scintigraphy (WBBPS). Int Angiol 23:355-367, 2004. 24. Lee BB, Mattassi R, Kim BT, et al. Advanced management of arteriovenous shunting malformation (AVM) with transarterial lung perfusion scintigraphy (TLPS) for follow-up assessment. Int Angiol 24:173-184, 2005. 25. Berwald C, Salazard B, Bardot J, et al. [Port wine stains or capillary malformations: surgical treatment] Ann Chir Plast Esthet 51:369-372, 2006. 26. Goldman MP, Fitzpatrick RE, Ruiz-Esparza J. Treatment of port-wine stains (capillary malformation) with the flashlamp-pumped pulsed dye laser. J Pediatr 122:71-77, 1993. 27. Lee BB. Klippel-Trenaunay syndrome: is this term still worthy to use? Acta Phlebologica 13:83-85, 2012. 28. Lee BB, Laredo J, Neville R, et al. Primary lymphedema and Klippel-Trenaunay syndrome. In Lee BB, Bergan J, Rockson S, ed. Lymphedema: A Concise Compendium of Theory and Practice. London: Springer Verlag, 2011. 29. Ziyeh S, Spreer J, Rossler J, et al. Parkes Weber or Klippel-Trenaunay syndrome? Noninvasive diagnosis with MR projection angiography. Eur Radiol 14:2025-2029, 2004. 30. Courivaud D, Delerue A, Delerue C, et al. Familial case of Parkes Weber syndrome. Ann Dermatol Venereol 133(5 Pt 1):445-447, 2006. 31. Schweigere L, Fotsis T. Angiogenesis and angiogenesis inhibitors in paediatric diseases. Eur J Pediatr 151:472-476, 1992.

K23346_Neligan_12_Pathophysiology_r3_0175-0188.indd 184

5/26/2015 9:02:39 AM

Chapter 12  Pathophysiology of Primary Lymphedema

185

32. Enciso JM, Hirschi KK. Understanding abnormalities in vascular specification and remodeling. Pediatrics 116:228-230, 2005. 33. Leu HJ. Pathoanatomy of congenital vascular malformations. In Belov S, Loose DA, Weber J, eds. Vascular Malformations. Reinbek, Germany: Einhorn-Presse Verlag, 1989. 34. Bastide G, Lefebvre D. Anatomy and organogenesis and vascular malformations. In Belov S, Loose DA, Weber J, eds. Vascular Malformations. Reinbek, Germany: Einhorn-Presse Verlag, 1989. 35. Alitalo K, Tammela T, Petrova TV. Lymphangiogenesis in development and human disease. Nature 438:946-953, 2005. 36. Maby-El Hajjami H, Petrova TV. Developmental and pathological lymphangiogenesis: from models to human disease. Histochem Cell Biol 130:1063-1078, 2008. 37. Karpanen T, Mäkinen T. Regulation of lymphangiogenesis—from cell fate determination to vessel remodeling. Exp Cell Res 312:575-583, 2006. 38. Rutkowski JM, Moya M, Johannes J, et al. Secondary lymphedema in the mouse tail: lymphatic hyperplasia, VEGF-C upregulation, and the protective role of MMP-9. Microvasc Res 72:161-171, 2006. 39. Rutkowski JM, Boardman KC, Swartz MA. Characterization of lymphangiogenesis in a model of adult skin regeneration. Am J Physiol Heart Circ Physiol 291:H1402-H1410, 2006. 40. Goldman J, Le TX, Skobe M, et al. Overexpression of VEGF-C causes transient lymphatic hyperplasia but not increased lymphangiogenesis in regenerating skin. Circ Res 96:1193-1199, 2005. 41. Leu HJ. Pathomorphology of vascular malformations: analysis of 310 cases. Int Angiol 9:147-155, 1990. 42. Woolard HH. The development of the principal arterial stems in the forelimb of the pig. Contrib Embryol 14:139-154, 1922. 43. Belov ST. Classification of congenital vascular defects. Int Angiol 9:141-146, 1990. 44. Belov ST. Anatomopathological classification of congenital vascular defects. Semin Vasc Surg 6:219224, 1993. 45. Borisov AV, Petrenko VM. The development of the thoracic duct lymphangions in the prenatal period of human ontogeny. Ontogenez 23:254-259, 1992. 46. Krutsiak VN, Polianskiĭ IIu. [Development of the thoracic duct in the prenatal period of human ontogeny] Arkh Anat Gistol Embriol 85:79-84, 1983. 47. Gloviczki P, Duncan AA, Kalra M, et al. Vascular malformations: an update. Perspect Vasc Surg Endovasc Ther 21:133-148, 2009. 48. Lee BB, Laredo J. Classification of congenital vascular malformations: the last challenge for congenital vascular malformations. Phlebology 27:267-269, 2012. 49. Lee BB, Laredo J, Neville R. Primary lymphedema as a truncular lymphatic malformation. In Lee BB, Bergan J, Rockson SG, eds. Lymphedema: A Concise Compendium of Theory and Practice. London: Springer Verlag, 2011. 50. Papendieck CM. Lymphangiomatosis and dermoepidermal disturbances of lymphangio-adenodysplasias. Lymphology 35:478-485, 2002. 51. Witte MH, Erickson R, Bernas M, et al. Phenotypic and genotypic heterogeneity in familial Milroy lymphedema. Lymphology 31:145-155, 1998. 52. Brice G, Child AH, Evans A, et al. Milroy disease and the VEGFR-3 mutation phenotype. J Med Genet 42:98-102, 2005. 53. Erickson RP. Lymphedema-distichiasis and FOXC2 gene mutations. Lymphology 34:1-5, 2001. 54. Brice G, Mansour S, Bell R, et al. Analysis of the phenotypic abnormalities in lymphoedema-distichiasis syndrome in 74 patients with FOXC2 mutations or linkage to 16q24. J Med Genet 39:478-483, 2002. 55. Burnand KG, Mortimer PS. Lymphangiogenesis and genetics of lymphoedema. In Browse N, Burnand KG, Mortimer PS, eds. Diseases of the Lymphatics. London: Arnold, 2003. 56. Witte MH, Erickson R, Reiser FA, et al. Genetic alterations in lymphedema. Phlebolymphology 16:1925, 1997. 57. Michelini S, Degiorgio D, Cestari M, et al. Clinical and genetic study of 46 Italian patients with primary lymphedema. Lymphology 45:3-12, 2012.

K23346_Neligan_12_Pathophysiology_r3_0175-0188.indd 185

5/26/2015 9:02:39 AM

186

Part III  Pathophysiology and Clinical Presentation

58. Bernas MJ, Witte CL, Witte MH. The diagnosis and treatment of peripheral lymphedema. Lymphology 34:84-91, 2001. 59. Kinmonth JB, Wolfe JH. Fibrosis in the lymph nodes in primary lymphoedema. Histological and clinical studies in 74 patients with lower-limb oedema. Ann R Coll Surg Engl 62:344-354, 1980. 60. Vercellio, Baraldini V, Coletti M, et al. Strategies in early management of pure lymphatic malformations. Lymphology 35:472-477, 2002. 61. Rosbotham JL, Brice GW, Child AH, et al. Distichiasis-lymphoedema: clinical features, venous function and lymphoscintigraphy. Br J Dermatol 142:148-152, 2000. 62. Northrup KA, Witte MH, Witte CL. Syndromic classification of hereditary lymphedema. Lymphology 36:162-189, 2003. 63. Greenlee R, Hoyme H, Witte M, et al. Developmental disorders of the lymphatic system. Lymphology 26:156-168, 1993. 64. Aagenaes O, van der Hagen CB, Refsum S. Hereditary recurrent intrahepatic cholestasis from birth. Arch Dis Child 43:646-657, 1968. 65. Lee BB, Laredo J, Seo JM, et al. Treatment of lymphatic malformations. In Mattassi R, Loose DA, Vaghi M, eds. Hemangiomas and Vascular Malformations. Milan: Springer-Verlag Italia, 2009. 66. Ferrell RE, Levinson KL, Esman JH, et al. Hereditary lymphedema: evidence for linkage and genetic heterogeneity. Hum Mol Genet 7:2073-2078, 1998. 67. Ferrell RE, Baty CJ, Kimak MA, et al. GJC2 missense mutations cause human lymphedema. Am J Hum Genet 86:943-948, 2010. 68. Ostergaard P, Simpson MA, Connell FC, et al. Mutations in GATA2 cause primary lymphedema associated with a predisposition to acute myeloid leukemia (Emberger syndrome). Nat Genet 43:929-931, 2011. 69. Evans AL, Bell R, Brice G, et al. Identification of eight novel VEGFR-3 mutations in families with primary congenital lymphoedema. J Med Genet 40:697-703, 2003. 70. Oh SJ, Jeltsch MM, Birkenhäger R, et al. VEGF and VEGF-C: specific induction of angiogenesis and lymphangiogenesis in the differentiated avian chorioallantoic membrane. Dev Biol 188:96-109, 1997. 71. Leak LV, Jones M. Lymphangiogenesis in vitro: formation of lymphatic capillary-like channels from confluent monolayers of lymphatic endothelial cells. In Vitro Cell Dev Biol Anim 30:512-518, 1994. 72. Jussila L, Alitalo K. Vascular growth factors and lymphangiogenesis. Physiol Rev 82:673-700, 2002. 73. Karkkainen MJ, Haiko P, Sainio K, et al. Vascular endothelial growth factor C is required for sprouting of the first lymphatic vessels from embryonic veins. Nat Immunol 5:74-80, 2004. 74. Morisada T, Oike Y, Yamada Y, et al. Angiopoietin-1 promotes LYVE-1-positive lymphatic vessel formation. Blood 105:4649-4656, 2005. 75. Yoon YS, Murayama T, Gravereaux E, et al. VEGF-C gene therapy augments postnatal lymphangiogenesis and ameliorates secondary lymphedema. J Clin Invest 111:717-725, 2003. 76. Bernier-Buzzanga J, Su WP. Noonan’s syndrome with extensive verrucae. Cutis 46:242-246, 1990. 77. Wyre HW Jr. Cutaneous manifestations of Noonan’s syndrome. Arch Dermatol 114:929-930, 1978. 78. Lanning P, Simila S, Suramo I, et al. Lymphatic abnormalities in Noonan’s syndrome. Pediatr Radiol 7:106-109, 1978. 79. Perry HD, Cossari AJ. Chronic lymphangiectasis in Turner’s syndrome. Br J Ophthalmol 70:396-399, 1986. 80. Oliver G. Lymphatic vasculature development. Nat Rev Immunol 4:35-45, 2004. 81. Rockson RG. Physiology, pathophysiology, and lymphodynamics. General overview. In Lee BB, Bergan J, Rockson SG, eds. Lymphedema: A Concise Compendium of Theory and Practice. London: SpringerVerlag, 2011. 82. Smoke A, Delegge MH. Chyle leaks: consensus on management? Nutr Clin Pract 23:529-532, 2008. 83. Bibby AC, Maskell NA. Nutritional management in chyle leaks and chylous effusions. Br J Community Nurs 19(Suppl 10):S6-S8, 2014.

K23346_Neligan_12_Pathophysiology_r3_0175-0188.indd 186

5/26/2015 9:02:39 AM

Chapter 12  Pathophysiology of Primary Lymphedema

187

84. Szuba A, Shin WS, Strauss HW, et al. The third circulation: radionuclide lymphoscintigraphy in the evaluation of lymphedema. J Nucl Med 44:43-57, 2003. 85. Brautigam P, Földi E, Schaiper I, et al. Analysis of lymphatic drainage in various forms of leg edema using two compartment lymphoscintigraphy. Lymphology 31:43-55, 1998. 86. Armenio S, Cetta F, Tanzini G, et al. Spontaneous contractility in the human lymph vessels. Lymphology 14:173-178, 1981. 87. Sjöberg T, Norgren L, Steen S. Contractility of human leg lymphatics during exercise before and after indomethacin. Lymphology 22:186-193, 1989. 88. Olszewski WL. Lymph vessel contractility. In Olszewski WL, ed. Lymph Stasis: Pathophysiology, Diagnosis and Therapy. Boca Raton, FL: CRC Press, 1991. 89. Olszewski WL, Engeset A. Intrinsic contractility of prenodal lymph vessels and lymph flow in human leg. Am J Physiol 239:775-783, 1980. 90. Reddy NP, Patel K. A mathematical model of flow through the terminal lymphatics. Med Eng Phys 17:134-140, 1995. 91. Olszewski WL. Contractility patterns of normal and pathologically changed human lymphatics. Ann NY Acad Sci 979:52-63, 2002. 92. Olszewski WL. Contractility patterns of human leg lymphatics in various stages of obstructive lymphedema. Ann NY Acad Sci 1131:110-118, 2008. 93. Olszewski WL. Physiology–lymph flow. In Lee BB, Bergan J, Rockson SG, eds. Lymphedema: A Concise Compendium of Theory and Practice. London: Springer-Verlag, 2011. 94. Szuba A, Rockson SG. Lymphedema: anatomy, physiology and pathogenesis. Vasc Med 2:321-326, 1997. 95. Olszewski WL. Physiology, biology, and lymph biochemistry. In Lee BB, Bergan J, Rockson SG, eds. Lymphedema: A Concise Compendium of Theory and Practice. London: Springer-Verlag, 2011. 96. Olszewski WL. Pathology and histochemistry. In Lee BB, Bergan J, Rockson SG, eds. Lymphedema: A Concise Compendium of Theory and Practice. London: Springer-Verlag, 2011. 97. Olszewski WL, Grzelak I, Engeset A. Cells in lymph draining normal human skin. Lymphology 15:168173, 1982. 98. Olszewski WL. Cells in lymph. In Olszewski WL, ed. Lymph Stasis: Pathophysiology, Diagnosis and Treatment. Boca Raton, FL: CRC Press, 1991. 99. Olszewski WL, Grzelak I, Ziolkowska A, et al. Immune cell traffic from blood through the normal human skin to lymphatics. Clin Dermatol 13:473-483, 1995.

K23346_Neligan_12_Pathophysiology_r3_0175-0188.indd 187

5/26/2015 9:02:39 AM

K23346_Neligan_12_Pathophysiology_r3_0175-0188.indd 188

5/26/2015 9:02:39 AM

C hapter 13 Pathophysiology of Secondary Lymphedema Etelka Földi

P

K ey P oints • The transport capacity of the lymphatic drainage system may be reduced as a result of surgery and/or radiotherapy for malignant tumors or after severe accidents, subsequently leading to lymphedema. • The pathophysiology and pathomorphology of secondary lymphedema include the degree of damage, stage of the disease, and presence of comorbidities.

Path

• Recognition of the stage of lymphedema is crucial to early treatment, whether surgical or conservative, to normalize the disturbed homeostasis.

Secondary lymphedema occurs after damage to the lymphatic drainage system and is most commonly associated with oncologic treatment (for example, after diagnostic or therapeutic lymphadenectomies or radiotherapy). Secondary lymphedema may also be a sign of malignant disease. Extensive soft tissue trauma and chronic inflammatory processes are the next most common causes of secondary lymphedema. The most commonly affected areas are the limbs; the least common are the head, trunk, and/or genitals. The lymphatic drainage system comprises the lymph capillaries (initial lymph vessels), precollectors, prenodal and postnodal collectors, and lymph trunks.1 The main tasks of the lymphatic drainage systems are as follows2,3: • Regulation of interstitial fluid volume • Maintenance of the normal metabolism of cells and extracellular matrix (ECM) • Removal of waste products • Ensure the circulation of lymphocytes • Migration of tissue macrophages, dendritic cells, and so forth • Elimination of cellular debris, including chemical components, such as inflammatory mediators, from injured tissue 189

K23349_Neligan_13_Pthophysiology_r4_0189-0194.indd 189

5/27/2015 9:27:22 AM

190

Part III  Pathophysiology and Clinical Presentation

When the physiologic function of the lymphatic drainage system is damaged, the homeostasis of the interstitium becomes disturbed both in the cellular and extracellular compartments. The disturbances of the complex relationships between the cellular compartment (fibroblasts, lymphocytes, macrophages, mast cells, and vascular and neurogenic structures) and ECM (collagen, glycoprotein, protein-glycan, glycosaminoglycans such as hyaluronan, chondroitin sulfate B, heparan, keratan sulfate, and other humoral substances and enzymes) lead to the remodeling of skin and subcutaneous tissues. Another important aspect is the influence on the immune system as a consequence of the impaired circulation of the lymphocytes.4

Pathophysiology and Pathomorphology The pathophysiology and pathomorphology of the secondary damaged lymphatic drainage system depend on several factors: • Degree of damage • Stage of the disease • Presence of comorbidities

Degree of Damage Acute Lymphedema Acute lymphedema is a rare condition that occurs most often after radical surgery (predominantly for pelvic malignancy) or severe accidents with soft tissue damage. The degree of damage to the lymphatic system can be so extensive that only a decreased number of functional lymph vessels remain. Their ability to regenerate is exceeded, and transport capacity in the lymphatic drainage system is dramatically reduced, with the consequence that lymphedema occurs and remains manifest.

Stage of the Disease Clinical Staging of Secondary Lymphedema In clinical practice, the stages of lymphedema are defined by a physical examination of the extremities and staged according to the parameters outlined in Box 13-1.

BOX 13-1  Staging of Lymphedema Latency: There is reduced transport capacity, with no clinical signs of swelling. Stage 1: Pitting edema subsides with limb elevation. Stage 2: Elevation of the limb is barely effective; in addition to pitting edema, there is hardening of the tissue resulting from fibrosis. Stage 3: Lymphostatic elephantiasis, or large volume of the limbs, appears in either column or lobular form. Attendant symptoms are congestive dermatitis, trophic skin changes, and fat deposition.

K23349_Neligan_13_Pthophysiology_r4_0189-0194.indd 190

5/27/2015 9:27:22 AM

Chapter 13  Pathophysiology of Secondary Lymphedema

191

Pathophysiologic Staging of Lymphedema Latency If the damage is less severe, sufficient numbers of functional lymph vessels remain and cope with the lymphatic load despite the reduced transport capacity. This condition refers to a latent or subclinical stage of lymphedema, in which swelling is not present despite the impaired lymph transport capacity. This stage of latency is characterized by increased lymph transport in the remaining functional lymph vessels, resulting from the activation of intrinsic factors. Stretching of the lymph vessel walls leads to increased lymphangion pulsation.5 Over time, these compensatory mechanisms become insufficient. Tissue fluid begins to accumulate, with pathologic changes to the lymph vessels themselves and the connective tissue. The transport capacity of the lymphatic drainage system is further compromised and lymphedema occurs. The latency stage can last for several months to years. Stage 1  In stage 1, the examination of tissue samples under a light microscope reveals indirect signs of raised fluid content. Tissue samples are transparent and can only be lightly stained with hematoxylin-eosin and Giemsa. The electron microscope image of stage 1 lymphedema is characterized by dislocation and compartmentalization of collagen fibers. Immunohistologic changes in stage 1 are rarely mentioned in the literature, but it must be assumed that the ECM reacts in a multitude of ways to the increased tissue fluid load. It can also be assumed that glycosaminoglycans (hyaluronic acids) play an important role in the deposition of lymph fluid.1,6 Stages 2 and 3  Pathomorphologic and pathophysiologic alterations in stages 2 and 3 of lymphedema are shown in Table 13-1. Examination of tissues under a light microscope reveals a high density of tissue through raised fiber content and increased cellular density, which surrounds the nerves, blood and lymph vessels, smooth muscle fibers, and fascia. Elastic fibers in particular remain in fragmented or granular forms. Furthermore, the inflammatory tissue changes lead to an increased deposition of adipose tissue. In chronic lymphedema of the skin, the dermis becomes

TABLE 13-1  Pathomorphologic and Pathophysiological Alterations in Stages 2 and 3 of Lymphedema Lymphedema Stages 2 and 3

Alteration

Collagen types I and III

Increased and altered

Fatty tissue

Increased

Dermoepithelial basal membrane

Discontinued

Cellular compartment of interstitium

CD4-lymphocytes ↑ Macrophages (M2 prominent) ↑ Mast cells ↑ Plasma cells ↑ Fibroblasts ↑ Dendritic cells ↑

Proinflammatory cytokine expression

Increased (TNF alpha; IL-1 beta; IL-6; VEGT-C; TGF beta)

IL-1 beta, Interleukin-1 beta; IL-6, interleukin-6; TGF beta, tumor growth factor beta; TNF alpha, tumor necrosis factor alpha; ↑, increased.

K23349_Neligan_13_Pthophysiology_r4_0189-0194.indd 191

5/27/2015 9:27:22 AM

192

Part III  Pathophysiology and Clinical Presentation

thicker, extending cones of tissue in a wartlike manner not only toward the surface of the skin, but also into the subcutaneous adipose tissue structures. Collagen and lipid deposition lead, among others, to alterations in the hydraulic conductivity of the tissue.7-9 It is unknown which factors in secondary lymphedema determine whether collagen fibrils will increase and thus predominantly lead to epifascial tissue fibrosis, including the deep fascia, with hardening of the skin and subcutis, or whether the deposition of fatty tissue will dominate, leading to a more voluminous lymphedema remaining soft on palpation. Based on my own animal experiments on chronic lymphedema in rabbits and laboratory rats, I found that 28 to 30 days after surgery, midgrade epifascial fibrosis with consecutive hardening of the skin and subcutis occurred in 50% of the animals, extreme fibrotic processes with severe hardening of the fascia occurred in 17%, and fatty deposition in the tissue was present in 33% of the animals. I had similar findings in human patients with secondary lymphedema based on clinical and ultrasound examinations, particularly with computed tomography.10 I suspect that gender plays a role (and subsequent hormonal influences). The nutritional status of the patient also influences the pathophysiology of lymphedema.

Presence of Comorbidities The role of comorbidities in secondary lymphedema has not been extensively researched. Each year we treat more than 1500 patients with secondary lymphedema at the Földi Clinic, most commonly after treatment for cancer, but only 19% of these patients have no other comorbidities. Blood capillaries, lymph capillaries, and the interstitium form a functional unit. Diseases that lead to increased permeability of the blood capillaries also influence the pathophysiology of lymphedema,11-13 and inflammatory processes can also exacerbate the disease. They play a role in the length of the so-called stage of latency, according to the International Society of Lymphology classification, and influence the severity of the damage to the lymphatic drainage system and the progression of lymphedema. Hormonal imbalances, such as polycystic ovarian syndrome,14 autoimmune processes15 (various forms of vasculitis), metabolic diseases16 (insulin resistance in type 2 diabetes), and neuropathies,17 are the most common according to our observations. There is a considerable shortage in research results in the literature compared with clinical data on possible changes to the pathomorphology and pathophysiology of secondary lymphedema with comorbidities. However, comorbidities play an important role in the treatment of lymphedema and the medical indications for conservative and/or surgical treatment of secondary lymphedema. It is generally known, for example, that in the combination form of secondary lymphedema with diabetes mellitus type 2 and polyneuropathy, not only is the transport capacity of the lymphatic drainage system reduced, but also the lymphatic load is elevated because of the increased permeability of blood capillaries.

Conclusion The transport capacity of the lymphatic drainage system may be reduced as a result of surgery and/or radiotherapy for malignant tumors or after severe accidents, and subsequently lymphedema may occur. The manifestation and progression of the swelling depend on the degree of damage and presence of accompanying diseases. Clinicians treating lymphedema must recognize

K23349_Neligan_13_Pthophysiology_r4_0189-0194.indd 192

5/27/2015 9:27:22 AM

Chapter 13  Pathophysiology of Secondary Lymphedema

193

that lymphedema is more than the simple accumulation of fluid in tissue. Lymphostasis leads to inflammatory processes and structural tissue remodeling. Treatment, either conservative or surgical, must be started as early as possible to normalize the disturbed homeostasis. The treatment of lymphedema is multifactorial. The recognition of the stage of lymphedema is important, and the patient must be optimized nutritionally and physically to achieve the optimal outcome.

C linical P earls • The understanding of the pathophysiology of secondary lymphedema is essential for the optimal management of the disease in clinical practice. • The quantitative relationship between the degree of damage to the lymphatic system and the amount of lymphatic load can already be estimated with a detailed medical history and clinical examination, allowing the clinician to adequately determine subsequent therapeutic measures. • The degree of required therapy also increases with comorbidities. • Secondary lymphedema is a chronic disease, and therefore teaching of guidelines for suitable lifestyle adaptations and self-treatment is highly important to patients, in addition to conservative and/or surgical treatment.

R EFERENCES 1. Földi M, Földi E. Földi’s Textbook of Lymphology, ed 3. Munich, Germany: Elsevier, 2012. 2. Olszewski WL. The pathophysiology of lymphedema—2012. Handchir Mikrochir Plast Chir 44:322328, 2012. 3. Olszewski WL, Jain P, Ambujam G, et al. Topography of accumulation of stagnant lymph and tissue fluid in soft tissues of human lymphedematous lower limbs. Lymphat Res Biol 7:239-245, 2009. 4. Alitalo K. The lymphatic vasculature in disease. Nat Med 17:1371-1380, 2011. 5. Zawieja DC. Contractile physiology of lymphatics. Lymphat Res Biol 7:87-96, 2009. 6. Roberts MA, Mendez U, Gilbert RJ, et al. Increased hyaluronan expression at distinct time points in acute lymphedema. Lymphat Res Biol 10:122-128, 2012. 7. Rutkowski JM, Markhus CE, Gyenge CC, et al. Dermal collagen and lipid deposition correlate with tissue swelling and hydraulic conductivity in murine primary lymphedema. Am J Pathol 176:1122-1129, 2010. 8. Ji RC. Lymphatic endothelial cells, lymphedematous lymphangiogenesis, and molecular control of edema formation. Lymphat Res Biol 6:123-137, 2008. 9. Kim SH, Turnbull J, Guimond S. Extracellular matrix and cell signaling: the dynamic cooperation of integrin, proteoglycan and growth factor receptor. J Endocrinol 209:139-151, 2011. 10. Földi E. Das Lymphödem: die Brücke vom Experiment zur klinik. Dokumenta Angiologorum, vol XXXI. Phlebologie 4:5-10, 2006. 11. Chappell D, Jacob M, Becker BF, et al. [Expedition glycocalyx: a newly discovered “Great Barrier Reef ”] Anaesthesist 57:959-969, 2008. 12. Cloutier N, Paré A, Farndale RW, et al. Platelets can enhance vascular permeability. Blood 120:13341343, 2012.

K23349_Neligan_13_Pthophysiology_r4_0189-0194.indd 193

5/27/2015 9:27:22 AM

194

Part III  Pathophysiology and Clinical Presentation

13. Eskens BJ, Zuurbier CJ, van Haare J, et al. Effects of two weeks of metformin treatment on whole-body glycocalyx barrier properties in db/db mice. Cardiovasc Diabetol 12:175, 2013. 14. Gluszak O, Stopińska-Gluszak U, Glinicki P, et al. Phenotype and metabolic disorders in polycystic ovary syndrome. ISRN Endocrinology 2012:569862, 2012. 15. Kukolnikova EL, Lapina NV. [Etiology and pathogenesis of secondary edema of the lower extremities in patients with limited pretibial myxedema on the background of hyperthyroidism] Khirurgiia (Mosk) 9:67-70, 2011. 16. Woodcock TE, Woodcock TM. Revised Starling equation and the glycocalyx model of transvascular fluid exchange: an improved paradigm for prescribing intravenous fluid therapy. Br J Anaesth 108:384394, 2012. 17. Vignes S, Lebrun-Vignes B. Sclerodermiform aspect of arm lymphoedema after treatment with docetaxel for breast cancer. J Eur Acad Dermatol Venereol 21:1131-1133, 2007.

K23349_Neligan_13_Pthophysiology_r4_0189-0194.indd 194

5/27/2015 9:27:23 AM

C hapter 14 Dermatologic Implications of Secondary Lymphedema of the Lower Leg Terence Ryan, S.R. Narahari, B. Vijaya, Madhur Guruprasad Aggithaya

K ey P oints • Peau d’orange skin texture, a common presentation of lymphedema, results from swelling of the upper dermis limited by tethering of adnexal structures to the skin. • Most dermatologic manifestations of lymphedema occur in the lower extremity.

P

• Venous insufficiency should be estimated in all patients with lymphedema. • Lymphatic filariasis is the most common cause of lower extremity lymphedema. It is transmitted by mosquito and is endemic in 78 developing nations. • Podoconiosis (or mossy foot), which results from damage to the tissues and lymph nodes of individuals who walk barefoot in irritant soil, is commonly observed in Ethiopia.

Der the

• Recurrent secondary infection from bacteria and fungi worsen lymphedema. • Excess collagen and dilated thick-walled lymphatics and loss of elastin are the key histologic changes associated with worsening lymphedema.

Most of the changes seen with lymphedema occur in the lower extremity rather than in the upper extremity. For that reason, this chapter concentrates on changes of the lower leg. Primary lymphedema is caused by congenital hypoplasia of the lymphatics, failures of connection between the initial and connecting lymphatics, or impaired connections with the blood vascular system. Lymph is intercellular fluid formed by blood capillaries plus macromolecules and cells generated in the skin but not taken up by the venous system. The classic theory is that 90% of this fluid is absorbed by the venules, but this is no longer considered the case. The new perspective on fluid exchange postulates that there is dwindling filtration along the arterial and venous blood capillaries. Thus the lymphatic system is the only effective means of removing capillary filtrate from most tissues.1 The brain is the most studied exception.

195

K23346_Neligan_14_Dermatologic Implicat_r3_0195-0214.indd 195

5/26/2015 9:03:55 AM

196

Part III  Pathophysiology and Clinical Presentation

If the balance between filtration and resorption is disturbed, fluid accumulates in the intercellular space and edema develops. When this becomes chronic, the tissues react with the development of fibrosis and hyperplasia—features of lymphedema—and in its severest form elephantiasis. Many causes account for the failure of normal function, including cardiac failure, renal failure, gravitational overload of the venous system and immobility, inflammation, cancer and its treatment by surgery or radiotherapy2 and lymphatic filariasis3 (see Chapter 15).

Normal and Pathologic Tissue Fluids There is, of course, always some fluid in the interstitium provided by its blood supply and controlled by Starling’s law that describes the balance between intravascular and tissue hydrostatic and oncotic forces. It is currently believed that lymphedema results from a failure of lymphatic function. It rarely is a total failure; mostly it is a partial failure aggravated by overload from a failing venous system or inflammation causing transudation from the blood capillary bed. The richest capillary bed of the skin is found immediately deep to the epidermis, and the network of initial lymphatics lies immediately deep to the blood supply at the junction of the upper and middle dermis. When sectioned horizontally, the rete ridges of the epidermis provide a closely knit network. The capillary bed lies deep to this by less than 0.1 mm and has slightly wider mesh and hairpin-shaped capillary loops projecting into the papillae. Below that is the initial lymphatic network with a much wider mesh (up to 1.0 mm) lying at the junction of the upper and middermis normally without projections into the papillae. This layering was well illustrated by Kubik and Manestar4and can be reproduced and downloaded from Ikomi and Schmid-Schönbein.5 Ultrasonographic studies of the skin show very clearly the relative water content versus cellular and fibrous components of the skin. They reveal the relative density of the epidermis, the collagen fibers of the dermis, and the water-filled interstitium. Most of the water content is found in the upper dermis, where the capillary bed is the richest. Ultrasonographic findings also show an accumulation of water in the upper dermis after prolonged venous overload from the gravitational effects of dependency of the leg, and there is a reduction of water in the upper dermis after 2 hours of elevation.6 Ultrasound examination can show localized indentation of the upper dermis by the fingernail or with a matchstick to a depth of 0.3 mm to indent and disperse the upper dermal interstitial fluid. Light massage with circular movements has the same effect, whereas deeper kneading massage shifts deep dermal water often like a deep bruise, bringing it to the upper dermal lymphatics. A great increase in water content, such as when the lymphatic system fails and cannot cope with venous overload, results in the expansion of the upper dermis, but adnexal components, such as the hair follicle, prevent such expansion in their immediate vicinity. It gives the appearance of orange peel, known as peau d’orange (Fig. 14-1). When fluid accumulation increases, such as in cellulitis caused by streptococcal infections of the regions of lymphedema, the hair follicles are unable to prevent further expansion of the dermis. As a result, lymphedema becomes painful, warm, and swollen, and the superficial skin becomes red. As the edema increases because of active infection, the peau d’orange appearance can be seen at the expanding edges of cellulitis while more central tissues are destroyed (Fig. 14-2).

K23346_Neligan_14_Dermatologic Implicat_r3_0195-0214.indd 196

5/26/2015 9:03:55 AM

Chapter 14  Dermatologic Implications of Secondary Lymphedema of the Lower Leg

197

The epidermis and upper dermis are subject to morphologic findings ranging from a flat epidermis—such as in atrophy—to extreme outward projection of the papilla and inward projection of the epidermis, giving the picture of acanthosis or hyperplasia (Fig. 14-3). The capillaries in the dermal papillae elongate and coil. Mostly the initial lymphatics maintain their position at the junction of the upper dermis with the mid-dermis. One of the features of lymphatic failure resulting from blockage of the collecting ducts into which they drain is that the initial lymphatics, which have only a single overlapping layer of endothelium

FIG. 14-1  Peau d’orange is swelling of the upper dermis, preventing expansion of the hair follicles and sweat glands.

FIG. 14-2  Further gross expansion of the upper dermis causing peau d’orange seen at the edge of the lesion with greater disruption in the center of the lesion.

FIG. 14-3  Hematoxylin-eosin–stained section of lymphedematous skin showing hypertrophy of the epidermis, dilated lymphatics, and patchy inflammatory infiltrate.

K23346_Neligan_14_Dermatologic Implicat_r3_0195-0214.indd 197

5/26/2015 9:03:57 AM

198

Part III  Pathophysiology and Clinical Presentation

A

B

C

FIG. 14-4  A, Hematoxylin-eosin stain. Dermis is showing dilated lymphatics with proteinaceous fluid. Lymphatic close to the overlying epidermis. Excess collagen in between dilated lymphatics (green arrow). Black arrow is pointing to a valve. Red arrow shows the smooth muscle in the wall filled with a few red cells. They are high in the upper dermal papillae in this patient with lymphedema. B, A severely hyperkeratotic leg (elephantiasis nostras verrucosa). The entire papillae, often lying deep to a severely hyperkeratotic epidermis. C, After removal of the excess keratin, the surface has the appearance of multiple polyps that appear similar to the villi of the small intestine.

in healthy individuals, develop smooth muscle, and the intralymphatic pressure rises as contractions become stronger (Fig. 14-4, A). One effect of this raised intralymphatic capillary pressure is to encourage the lymphatics undergoing lymphangiogenesis to penetrate higher in the upper dermis and into the widened papilla. As this becomes a dominant feature, the papilla extends closer to the surface, and the dilated lymphatic may reach the surface (Fig. 14-4, B). The entire papillae may resemble small intestinal villi (Fig. 14-4, C). Capillaries and lymphatics extend to the peak of each villus or present as vesicular fluid-filled lymphatics (see Fig. 14-6). Histologically there may be a smooth muscle–lined deep cyst into which the lymphatics drain and receive their lymph under high pressure.7 The variations in the dimensions of the polyplike protrusions can be observed in Fig. 14-5. The cobblestone-like nodules are often referred to as elephantiasis nostras verrucosa.8 Originally the Latin term for “ours,” nostras, referred to belonging to “our region,” indicating its typically tropical prevalence.

K23346_Neligan_14_Dermatologic Implicat_r3_0195-0214.indd 198

5/26/2015 9:03:58 AM

Chapter 14  Dermatologic Implications of Secondary Lymphedema of the Lower Leg

199

B

A

C

D

E

FIG. 14-5  A and B, As the tissues expand in lymphedema, the polypoid dimensions become larger. Individually they are frequently filled with compressible edema and dilated lymphatics, making them indentable and compressible, unlike the equally common hard nodules of fibrous tissue. C, A single large nodule composed of noncompressible whorls of collagen fibers. D, Scattered nodules of whorls of collagen. E, Numerous nodules of collagen, but at the same time the forefoot also shows much smaller surface changes, which are described as “mossy.”

The so-called mossy foot is now also used to describe podoconiosis, which is caused by damage to the tissues and lymph nodes by an alkaline colloid of silica from the soil penetrating the skin in individuals who walk barefoot; the condition is commonly observed in Ethiopia.9 Mossy foot, which is also described in Aboriginal Australians, usually begins around the toes. Gradually there is accompanying hypertrophy, and deep crevasses become entry points for bacteria and soil irritants. Lymph leaks and wets the surface of the skin, causing maceration, and secondary infection by bacteria and fungi occurs. The production of excess collagen gives rise to increasingly larger protuberances in which the lymphatics are sometimes hugely dilated. The condition is reported in all cases of lymphedema and occurs as a result of chronicity and repeated secondary infection of the area.

K23346_Neligan_14_Dermatologic Implicat_r3_0195-0214.indd 199

5/26/2015 9:04:00 AM

200

Part III  Pathophysiology and Clinical Presentation

A

B

C

D

FIG. 14-6  Lymphangioma circumscriptum. A, Dilated lymphatics close to the surface contain clear fluid. B, They also contain pink fluid resulting from a little blood contamination to deep red filled with blood. C, A milky white appearance occurs when the lymphatics are filled with lymph from the small intestine. D, A single leaking lymphatic is illustrated (arrow).

Lymphangioma Circumscriptum Dilated lymphatics often penetrate the papilla and epidermis to reach the surface of the skin. They develop contractile smooth muscle and loss of valves. Whimster7 found that these contractile lymphatics often drain into large high-pressure cystic dilations of the collecting lymphatics. The color of the superficial vesicular lymphatics depends on the number of red cells, or they can be milky white from chyle (Fig. 14-6). The term chylous reflux describes a backflow of chyle from its normal route from the bowel through the cisterna chyli and thoracic duct to reach the bloodstream in the great veins of the thorax. The clinical presentations include small blisters containing chyle-chylous vesicles, which may appear on the skin in the lower parts of the body and discharge milky fluid, thus forming one type of chylous fistula. Chylometrorrhagia causes vaginal discharge of chyle. Chyle may collect in the serous cavities of the body—producing chylothorax, chylous ascites, and chylocele.10

K23346_Neligan_14_Dermatologic Implicat_r3_0195-0214.indd 200

5/26/2015 9:04:01 AM

Chapter 14  Dermatologic Implications of Secondary Lymphedema of the Lower Leg

201

FIG. 14-7  Examples of late-stage elephantiasis and great hypertrophy observed in lymphedema.

A

B

FIG. 14-8  A, Hematoxylin-eosin–stained section of lymphedema demonstrating excess collagen and dilated thick-walled lymphatics. B, NB, a lymphatic draining into a filled and dilated muscular lymphatic.

Tissue Hypertrophy As well demonstrated in studies of experimental lymphedema in dogs,11 there is huge enlargement of the tissues in elephantiasis in the later stage of lymphedema12,13 (Fig. 14-7). However, enlargement of the tissues is only partially the result of the accumulation of water. Such accumulation is supposedly enough to create tissue expansion and provide mechanical stress to tissue fibroblasts, which respond by a massive increase in collagen, some adipose tissue, lymphangiogenesis, and angiogenesis (Fig. 14-8).

K23346_Neligan_14_Dermatologic Implicat_r3_0195-0214.indd 201

5/26/2015 9:04:02 AM

202

Part III  Pathophysiology and Clinical Presentation

A

B

FIG. 14-9  A, Negative Stemmer sign in a patient with Ehlers-Danlos syndrome. The skin over the toes is easily picked up. B, Positive Stemmer sign showing measuring resistance to picking up a skin fold.

Stemmer Sign The chronic stimulus to the fibroblast-raised interstitial hydrostatic pressure is first identified in the toes of tall young adults from the standing and sitting positions. As shown by a physical therapist in the Földi Clinic (personal communication), about 20% of young adults will have some stiffness of the skin, and thus the skin over the dorsum of the toes cannot be easily picked up between the finger and thumb (Fig. 14-9).

Elastin As Unna14 demonstrated in his 1896 textbook, the normal lymphatic vessel is supported by elastin (Fig. 14-10), and he showed that edema leads to a loss of elastin. Most commonly elastases from neutrophils or activated mast cells and macrophages are responsible. The destruction of elastin has been found in granulomatous tissue in leprosy.15 Fibrosis is a cause of the nonpitting status of lymphedema, which is also termed brawny edema. The pitting of venous edema is seen in Fig. 14-11. Deep indentation over a wider area requires much greater pressure in lymphedema. Localized indentation to a depth of 0.3 mm by a fingernail or thumb can be achieved easily in all types of edema. The pattern of lymphedema that produces great expansion of the papilla filled with a dilated lymphatic will be softer and more compressible nodules than that produced by fibrosis (see Fig. 14-5, A). They will indent if pressure is applied to only one nodule. Often these nodules contain dilated dermal lymphatic vessels. Collagen is often found in collections of tightly bound fibers or whorls producing nodules, which may be few or many. They are firm and not compressible. They are usually observed in sites of surgical debridement of lymphedematous tissue (Fig. 14-12).

K23346_Neligan_14_Dermatologic Implicat_r3_0195-0214.indd 202

5/26/2015 9:04:02 AM

Chapter 14  Dermatologic Implications of Secondary Lymphedema of the Lower Leg

203

B

A

C

D

FIG. 14-10  Orcein stain. A and B, Normal lymphatic supported by elastin. A, The lymphatic lies at the junction of the upper and middle dermis and is longer than blood capillaries at this level in the skin. B, The elastin surrounding the lymphatic has tangential fibers extending to the epidermis. C, There is an absence of elastin, but there is expansion of the upper dermis by angiogenesis. The epidermis is thinned. The initial lymphatics (arrows) are now deep and dilated. D, Higher magnification showing angiogenesis.

A

B

FIG. 14-11  A, Pitting edema. B, Pressure with the thumb is resisted by brawny edema.

K23346_Neligan_14_Dermatologic Implicat_r3_0195-0214.indd 203

5/26/2015 9:04:04 AM

204

Part III  Pathophysiology and Clinical Presentation

FIG. 14-12  Surgical removal of excess tissue and grafts taken from the thigh has resulted in keloid development at both sites.

A

B

FIG. 14-13  Two examples of fissuring of the epidermis, common in individuals who do not wear shoes and are agricultural workers. A, Heels. B, Plantar surface of the forefoot.

Common Entry Points for Bacteria and Irritant Soils The recognition of easy access through the epidermis for bacteria and irritants has led to an emphasis on skin care. It is common for fissures to occur on the soles of the feet of individuals who do not wear footwear (Fig. 14-13). It is a feature of the shoeless agricultural worker and an important contribution to podoconiosis in the irritant soil of Ethiopia.9 It is also a feature of middle-aged overweight women and the forefoot of atopic children.16 One of the best recognized entry points is an intertrigo between the toes (Fig. 14-14). Here moisture may collect and cause maceration. If there is no secondary contamination from fungi and

K23346_Neligan_14_Dermatologic Implicat_r3_0195-0214.indd 204

5/26/2015 9:04:05 AM

Chapter 14  Dermatologic Implications of Secondary Lymphedema of the Lower Leg

205

Candida, the maceration and scaling do not extend beyond the crevasse. Fungal contamination usually extends beyond the crevasse, and scaling is visible beyond the crevasse. If there is scale beyond the crevasse, then a candidal or fungal infection should probably be suspected, because they usually extend beyond the site of maceration. With the leg dependent, the depth of the crevasse may be difficult to examine, but it should be aerated by expanding with foam rubber (Fig. 14-15, A). After the edema is reduced by therapy, full examination of the depth of the crevasse is facilitated by elevation (Fig. 14-15, B). In advanced lymphedema in which volume becomes enormous, the crevasses are deep and multiple.

A

B

FIG. 14-14  A, Common entry point for bacteria or soil irritants is macerated skin. It is often confined deep in a crevasse, for example, between the toes. B, Another entry point is deep in a skinfold of hypertrophic skin.

B

A

FIG. 14-15  Two examples of opening up a deep crevasse for closer examination. A, By using pieces of foam. B, By elevation.

K23346_Neligan_14_Dermatologic Implicat_r3_0195-0214.indd 205

5/26/2015 9:04:07 AM

206

Part III  Pathophysiology and Clinical Presentation

Nail Changes Nail growth is disturbed by lymphedema. For unknown reasons, there is typically shortening and upward tilting of the nail, which is termed the ski jump nail (Fig. 14-16, A). In growing, the nail often shows hypertrophy of the tissue at the sides of the nail, and bandaging or tight-fitting shoes or bandages press the overgrown tissue into the nail (Fig. 14-16, B). There may also be gross onychogryphosis (Fig 14-16, C). Yellow nail syndrome (Fig. 14-17) is an unexplained condition in which nail growth ceases and the nail is thick and yellow. This syndrome is characterized by slowly growing, dystrophic, yellow nails, peripheral lymphedema, pleural effusions, rhinosinusitis, and bronchiectasis, with consecutive recurrent infections of the lower respiratory tract.17,18

A

FIG. 14-16  A, The elevated shape of the nail affected by lymphedema may resemble a ski jump. B, In growing, the nail shows hypertrophied skin lateral to the nail when compressed by shoes or bandages against the nail. C, The nail may be onychogryphotic.

B

C

FIG. 14-17  Rarely, nails stop growing and become uniformly discolored yellow. This is the yellow nail syndrome, which is associated with pleural effusions.

K23346_Neligan_14_Dermatologic Implicat_r3_0195-0214.indd 206

5/26/2015 9:04:08 AM

Chapter 14  Dermatologic Implications of Secondary Lymphedema of the Lower Leg

207

Adipose Tissue Stimulation of overgrowth of adipose tissue is a feature of lymphedema,11,19,20 but there are normally small collections of adipose tissue around hair follicles and sweat glands (Fig. 14-18). In lymphedema adipose cells and tissue produce smooth, rubbery feeling masses of tissue (Fig. 14-19). They are mostly seen in late stage II of International Society of Lymphology grades. Lipedema characteristically involves localized primary adipose hypertrophy with a secondary collection of fluid, sparing the feet, with relatively less fibrosis and fewer other signs of tissue

FIG. 14-18  Hematoxylin-eosin stain of a full-thickness skin biopsy. Adipose tissue is normally subcutaneous, but it often protects adnexal tissue, such as hair follicles and sweat glands, from shearing forces and may provide essential fatty acids.

FIG. 14-19  Pathology of lymphedema also stimulates growth and inflammation of adipose tissue. It gives rise to local, often smooth-skinned, rubbery, consistent swellings.

K23346_Neligan_14_Dermatologic Implicat_r3_0195-0214.indd 207

5/26/2015 9:04:09 AM

208

Part III  Pathophysiology and Clinical Presentation

A

B

C

D

FIG. 14-20  A, In lipedema. B, Hematoxylin-eosin–stained epidermis shows effacement of rete ridges. The dermis is expanded or thickened with dense collagen bundles. C, Dermis shows fragmented elastic fibers (in black Verhoeff–van Gieson stain). D, Subcutaneous tissue shows hypertrophic mature adipose cells.

component hypertrophy19-21 (Fig. 14-20, A). Some characteristics of this physical condition are deposition of excess fat on the legs (described classically as an Egyptian column shape) and arms with a negative Stemmer sign.22 The histopathologic findings of lipedema have not been extensively studied. In a few biopsied cases at the Institute of Applied Dermatology, Kasargod, India, the epidermis in lipedema was thin, elastic fibers were fragmented, and the dermis was thickened from collagen deposits (Fig. 14-20, B). Subcutaneous tissue showed hypertrophic adipose cells (Fig. 14-20, C). Others have emphasized inflammation, hypoxia, stem cells, and special features, such as an excess of CD68 macrophages and Ki671CD341cells (stem cells).23,24 It is difficult to manipulate by massage, because the forces applied are dissipated by adipocytes. It is easier to reshape the swollen adipose tissue than it is to reduce it (Fig. 14 -21).

K23346_Neligan_14_Dermatologic Implicat_r3_0195-0214.indd 208

5/26/2015 9:04:10 AM

Chapter 14  Dermatologic Implications of Secondary Lymphedema of the Lower Leg

A

209

B

FIG. 14-21  A, Lymphedema, mostly comprised of adipose tissue, was located below the knee before bandaging. B, After 25 days of compression, it is reshaped and covers the knee.

Venous Reflux The three main contributors to lymphedema are: 1. Failure of lymph flow into and along the initial lymphatics 2. From the initial lymphatic system preferentially and by fast track into the collecting system 3. By a smooth muscle pump along the collecting lymphatics and through the lymph nodes and finally through the thoracic duct into the great veins assisted by deep breathing Such failure is often only partial, but it is greatly exacerbated by inflammation resulting from the entry of bacteria or irritants, and as emphasized previously, by venous overload caused by venous reflux.6 Dilated veins should be recorded on standing and managed by elevation, ankle movements, and support bandages or hosiery. Ulceration is more often the result of venous failure than lymphatic failure. It is common for persons with “heavy legs” to sit with their legs dependent and immobile. This results in gravitational overfilling of the veins. In lymphedema, venous incompetence must be recognized on the observation that dilated veins are the most obvious when standing (Fig. 14-22). Venous reflux is the retrograde flow in a vein in response to a stimulus such as a calf squeeze. It occurs in the standing position when valves are incompetent. Leakage of the distended venous capillaries results in characteristic pigmentation caused by hemosiderin (Fig. 14-23, A). In chronic venous insufficiency, increased matrix turnover results in a tendency for ulceration through lipodermatosclerosis. It should be expected if there is any ulceration at or above the ankle when minor trauma does not heal, especially when there are long-standing untreated varicose veins. Diabetes should be suspected when ulceration of the toes or sole of the foot is present (Fig. 14-24). In all patients the surgeon should always inquire about medication history, such as oral contraceptives and antipsychotic drugs.

K23349_Neligan_14_Dermatologic Implicat_r3_0195-0214.indd 209

5/28/2015 8:19:32 AM

210

Part III  Pathophysiology and Clinical Presentation

FIG. 14-22  Venous overload with dilated varicose veins is aggravated by lymphedema. Heavy legs and the tendency to sit immobile with the legs dependent may be observed in the clinic.

A

B

FIG. 14-23  A, One feature is leakage of blood from dilated capillary beds with resulting hemosiderin pigmentation. B, Retention of melanin pigment in some ichthyotic patterns of hyperkeratosis. Pigmentation resulting from hemosiderin is not a feature of lymphedema.

FIG. 14-24  Ulceration at pressure points on the foot should lead to a check for diabetes.

K23346_Neligan_14_Dermatologic Implicat_r3_0195-0214.indd 210

5/26/2015 9:04:12 AM

Chapter 14  Dermatologic Implications of Secondary Lymphedema of the Lower Leg

211

Other Dermatologic Changes Folliculitis sometime occurs from bacteria but can also often be from irritants. One of these is the Ayurvedic oil used in massage, especially during the integrative treatment of lymphedema as practiced in India. Scarification marks are frequently from incisions made by traditional health practitioners to allow lymph to drain (Fig. 14-25). Ulcerated nodules resulting from malignancy (Fig. 14-26) are sometimes a consequence of lymphedema (for example, a lymphosarcoma named after Stewart and Treves) (Fig. 14-27). This sarcoma is a not too rare final consequence of lymphedema. It has been suggested that it is from impaired immunity, which is a feature of a failed lymphatic system. A squamous cell carcinoma of the foot reminds us that malignancies of every type may be more common when there is an impaired immune system.25-27

FIG. 14-25  Traditional healers cauterize lymphedema to drain the lymph. This harmful practice, which was observed until recently worldwide but now is mostly confined to rural Africa and India, results in acute inflammatory episodes resulting from secondary infection. Recurrent attacks worsen the lymphedema.

FIG. 14-26  Diffuse large B-cell type of non-Hodgkin lymphoma nodules and ulcerations developed over long-standing lower leg lymphedema of 13 years’ duration. Such nodules also occur from lymphangiosarcoma, a rare complication of lymphedema.

K23346_Neligan_14_Dermatologic Implicat_r3_0195-0214.indd 211

5/26/2015 9:04:13 AM

212

Part III  Pathophysiology and Clinical Presentation

FIG. 14-27  Erythematous and blood-stained nodules of lymphangiosarcoma developed over 7 years’ duration of lymphedema of the leg.

C linical P earls • In a patient with lymphedema, clinicians should look for features that contribute to disease progression, especially bacterial entry lesions. • When associated with nodules and ulcers, other causes and complications of lymphedema should be explored. • In all patients with lymphedema, the presence of reducible pitting edema and associated venous insufficiency should be estimated. • Lipedema is difficult to treat, and compression therapy helps to reshape the limb.

R EFERENCES 1. Levick JR, McHale N. The physiology of lymph production and propulsion. In Browse N, Burnand KG, Mortimer PS, eds. Diseases of the Lymphatics. London: Arnold, 2003. 2. Beesley V, Janda M, Eakin E, et al. Lymphedema after gynecological cancer treatment: prevalence, correlates, and supportive care needs. Cancer 109:2607-2614, 2007. 3. Michael E, Bundy DA, Grenfell BT. Re-assessing the global prevalence and distribution of lymphatic filariasis. Parasitology 112:409-428, 1996. 4. Kubik S, Manestar M. Anatomy of the lymph capillaries and pre-collectors of the skin. In Bollinger A, Partsch H, Wolfe JHN, eds. The Initial Lymphatics. New York: Thieme-Stratton, 1985. 5. Ikomi F, Schmid-Schönbein GW. Lymph transport in the skin. Clin Dermatol 13:419-427, 1995. 6. Hu D, Phan TT, Cherry GW, et al. Dermal edema assessed by high frequency ultrasound in venous leg ulcers. Br J Dermatol 138:815-820, 1998. 7. Whimster IM. The pathology of lymphangioma circumscriptum. Br J Dermatol 94:473-486, 1976. 8. Sha M. Images in clinical medicine: elephantiasis nostras verrucosa. N Engl J Med 370:2520, 2014.

K23346_Neligan_14_Dermatologic Implicat_r3_0195-0214.indd 212

5/26/2015 9:04:13 AM

Chapter 14  Dermatologic Implications of Secondary Lymphedema of the Lower Leg

213

9. Davey G, Tekola F, Newport MJ. Podoconiosis: non-infectious geochemical elephantiasis. Trans Roy Soc Trop Med Hyg 101:1175-1180, 2007. 10. Kinmonth JB, Taylor GW. Chylous reflux. Br Med J 1:529-532, 1964. 11. Casley-Smith JR, Clodius L, Piller NB. Tissue changes in chronic experimental lymphedema in dogs. Lymphology 13:130-141, 1980. 12. International Society of Lymphology. The diagnosis and treatment of peripheral lymphedema. 2009 Consensus Document of the International Society of Lymphology. Lymphology 42:51-60, 2009. 13. Bennuru S, Maldarelli G, Kumaraswami V, et al. Elevated levels of plasma angiogenic factors are associated with human lymphatic filarial infections. Am J Trop Med Hyg 83:884-890, 2010. 14. Unna PG. The Histopathology of the Diseases of the Skin. Edinburgh: Macmillan & Co, 1896. 15. Ryan TJ, Jones R, Mortimer PS, et al. Lymphatics in leprosy: relationship to elastic fibres and observations following intra-lesional injections of colloidal carbon. Lepr Rev 73:52-63, 2002. 16. Ashton RE, Griffiths WA. Juvenile plantar dermatosis—atopy or footwear? Clin Exp Dermatol 11:529534, 1986. 17. Norkild P, Kroman-Anderson H, Struve-Christensen E. Yellow nail syndrome—the triad of yellow nails, lymphedema and pleural effusion. A review of the literature and a case report. Acta Med Scand 219:221-227, 1986. 18. Hoque SR, Mansour S, Mortimer PS. Yellow nail syndrome: not a genetic disorder? Eleven new cases and a review of the literature. Br J Dermatol 156:1230-1234, 2007. 19. Ryan TJ. Lymphatics and adipose tissue. Clin Dermatol 13:493-498, 1995. 20. Földi M, Földi E, Kubic S. Textbook of Lymphology. Munich: Elsevier, 2007. 21. Reich-Schupke S, Altmeyer P, Stücker M. Thick legs— not always lipedema. J Dtsch Dermatol Ges 11:225-233, 2013. 22. Godoy GM, Barufi F, Godoy MF. Lipedema: is aesthetic cellulite an aggravating factor for limb perimeter? J Cutan Aesthet Surg 3:167-168, 2013. 23. Suga H, Araki J, Aoi N, et al. Adipose tissue remodeling in lipedema: adipocyte death and concurrent regeneration. J Cutan Pathol 36:1293-1298, 2009. 24. Szolnoky G, Kemény L. Lipoedema: from clinical presentation to therapy. Further aspects. Br J Dermatol 162:889, 2010. 25. Ruocco V, Astirata C, Guerera V, et al. Kaposi’s sarcoma on a lymphedematous immunocompromised limb. Int J Dermatol 23:56-60, 1984. 26. Mallon E, Powell SM, Mortimer PS. Contact sensitization and antigen recall in post-mastectomy lymphedema. Br J Dermatol 131(Suppl 44):14-29, 1994. 27. Ryan TJ, Mallon EC. Lymphatics and the processing of antigen. Clin Dermatol 13:485-492, 1995.

K23346_Neligan_14_Dermatologic Implicat_r3_0195-0214.indd 213

5/26/2015 9:04:13 AM

K23346_Neligan_14_Dermatologic Implicat_r3_0195-0214.indd 214

5/26/2015 9:04:13 AM

C hapter 15 Filaria Gurusamy Manokaran, Rajiv Agarwal, Devisha Agarwal

K ey P oints • Adenolymphangitis is an important clinical manifestation of lymphatic filariasis; recurrence contributes to the progress of the disease. • Meticulous compliance with nonsurgical methods of treatment is essential to avoid surgery or prevent recurrence after surgery. • A combination of physiologic surgery, such as a nodovenous shunt or a lymphaticovenous shunt, with an immediate reduction or debulking procedure with skin grafting is a very effective treatment method. • Lymphatic filariasis has no cure.

Fila Approximately 120 million people worldwide are at risk of contracting filariasis, and 70 million people have established filariasis, of whom 40 million have lymphedema. Medical and paramedical professionals and health planners need to understand this disease and attempt to prevent and decrease morbidity by various means, including surgery. The World Health Organization is working toward elimination of lymphatic filariasis by 2020. In the meantime, patients with clinical manifestations require lifelong treatment. In the future, earlier detection and treatment may reduce the severity of the disease but will not eliminate the problem. Lymphatic filariasis is one of the most incapacitating of chronic diseases. Previously, no treatment was thought possible. Ancient sculptures and scriptures depicted lymphatic filariasis of the lower limb; these are currently seen in many temples in India. According to Manusmriti (300 BC) from Hindu mythology, the disease was considered a result of karma, a retribution for actions committed in a previous life. Some primitive treatments were performed. As science and technology progressed, the organism and its mode of transmission from mosquitoes to humans were identified. Initially many medical and surgical treatments were attempted, but invariably they were unsuccessful, and lymphatic filariasis became a neglected tropical disease.

215

K23346_Neligan_15_Filaria_r3_0215-0234.indd 215

5/26/2015 9:09:39 AM

216

Part III  Pathophysiology and Clinical Presentation

Pathogenesis of Lymphatic Filariasis Lymphatic filariasis is a vector-borne disease of tropical countries caused by infection with filarial worms transmitted from the Culex species of mosquitoes (Fig. 15-1). The breeding of these mosquitoes is associated with aquatic plants such as Pistia stratiotes (water lettuce) and Salvinia auriculata (butterfly fern). The infective mosquitoes harbor the larvae (microfilariae), which enter the human host through bite wounds and migrate into the lymphatic system, where they develop into adult male and female worms. These worms—most commonly Wuchereria bancrofti (Fig. 15-2, A), Brugia malayi (Fig. 15-2, B), and Brugia timori (Fig. 15-2, C)—invade the lymphatics, leading to pathologic changes culminating in filarial disease manifestations.1 In mainland India, the causative nematodes are mostly W. bancrofti (see Fig. 15-2) and B. malayi, which are transmitted by specific genera of mosquitoes that include Culex quinquefasciatus, Anopheles, and others. B. malayi infection is now reportedly restricted to rural areas of South India.

MOSQUITO STAGES

HUMAN STAGES

1 An infected mosquito takes a blood meal. Infective L3 larvae enter the human skin through the bite wound and migrate to the lymphatic system.

L4

7 Between days 11 and 13, L2 larvae develop and molt into infective L3 stage. L3 larvae migrate through the hemocoel to enter the mosquito’s mouthparts.

6 Between days 6 and 10 after entering the mosquito, L1 microfilariae develop within the thoracic muscles into L2 “sausage” stage.

2 Between days 9 and 14 after entering the human host, L3 larvae transform into L4 larvae. Within 6 to 12 months, L4 larvae grow into mature adult worms.

Adults

L3 3 Adult worms nest in the lymphatic system and mate. The female worm releases thousands of sheathed microfilariae into lymphatic circulation, where they migrate to the bloodstream.

L2

5 The ingested microfilariae move to the mosquito’s stomach and shed their sheaths. They migrate through the stomach wall, bore between wing muscle fibers, and enter thoracic flight muscles. 4 A mosquito takes a blood meal from an infected individual and ingests L1 microfilariae.

L1

FIG. 15-1  Life cycle of a lymphatic filarial parasite.

K23346_Neligan_15_Filaria_r3_0215-0234.indd 216

5/26/2015 9:09:40 AM

217

Chapter 15  Filaria

A

B

C

FIG. 15-2  Worms known to cause lymphedema. A, W. bancrofti. B, B. malayi. C, B. timori.

FIG. 15-3  Characteristic histopathologic appearance of skin and subcutaneous tissues in chronic lymphatic filariasis. The tissue is lined by hyperkeratotic stratified squamous epithelium. The superficial dermis shows edema and sclerosis, along with proliferating small blood vessels and ill-defined epitheloid granulomas associated with lymphocytes, histiocytes, plasma cells, and occasional giant cells. Focal vasculitic lesions are also present.

The worms live in the lymph vessels and lymph nodes by making nests in the dilated lymphatics. The term worm nests refers to dilated lymphatic vessels with the characteristic movement pattern of worms, as evidenced on ultrasound examination. This early pathologic state predisposes the system to lymph dysfunction by causing incompetence of the unidirectional valves. Lymphatic filariasis is characterized by the presence of live adult parasites in the lymphatic system, with larvae (microfilariae) in the blood at certain times of the night (generally) in the early stage of the development of the lymphedema. The presence of microfilariae indicates an early stage in the life cycle in which the adult worms live in the lymph vessels and lymph nodes while they release microfilariae into the bloodstream of the host. The presence of microfilariae in the host bloodstream is called microfilaremia. This happens after the worms mate and the females produce millions of microfilariae, which migrate to the blood circulation. The sheathed microfilariae begin to appear in the blood circulation in 6 to 12 months after infection. They remain in the arterioles of the lungs during the day and emerge into the peripheral circulation at night. The periodicity of microfilariae coincides with the biting activity of the vector. They are taken up by blood-feeding vectors that spread the disease.

K23346_Neligan_15_Filaria_r3_0215-0234.indd 217

5/26/2015 9:09:40 AM

218

Part III  Pathophysiology and Clinical Presentation

Histopathologically, the filarial tissue shows hyperproliferation of keratinocytes, focal acantholysis, an accumulation of lymphocytes at the epidermodermal junction, and profuse pericapillary and perivenular mononuclear infiltrations (Fig. 15-3).

Manifestations of Lymphatic Filariasis The manifestations can be categorized as acute and chronic disease states (Table 15-1). Acute manifestations are comprised of adenolymphangitis, epididymoorchitis, and funiculitis, whereas chronic manifestations include lymphedema, hydrocele, elephantiasis, chyluria, chylothorax, chylascites, lymph scrotum, and tropical pulmonary eosinophilia. Advanced stages of lymphedema are characterized by increasing dilation and tortuosity of the lymphatics, endothelial proliferation, formation of new lymph channels, and obstructive changes with dermatosclerosis and warty lesions.2 Genital manifestations include filarial scrotum, ram’s horn penis, genital vesicles, and edema. Atypical lymphatic filariasis manifests as fleeting joint pains and lymphangitis (string sign). It can affect the breast, gluteal region, abdomen, and suprapubic region as isolated lesions. Manifestations are most commonly seen in the lower limb, more often in females.

TABLE 15-1  Clinical Presentation of Lymphatic Filariasis Clinical Stage

Clinical Feature

Pathogenesis

1  Asymptomatic parasite carrier state

Lymph vessel dilation Lymph vessel tortuosity

Subclinical infection

   a. Acute dermatoadenolymph­ angitis

Fever and chills Extremity warm and painful, swollen and tender Lymphangitis, cellulitis, abscess

Entry of bacteria and pathogens through the lesion

   b. Acute filarial lymphangitis

Tender lymph nodes Long tender cords underneath the skin

At site where adult worms die

   c. Acute epididymoorchitis and funiculitis

Pain, tenderness, and swelling of scrotum

Precipitated by secondary infection

  a. Lymphedema

Swelling of the extremity Three grades

Edema from lymphatic blockade

  b. Hydrocele

Swelling of scrotum

Accumulation of fluid in the tunica vaginalis

  c. Elephantiasis

Grade III lymphedema with dermatosclerosis and papillomatous lesions

Gross increase in edema

  d. Chyluria

Painless milky urine

Blockage of retroperitoneal nodes below cisterna chyli with reflux into renal lymphatics

   e. O  ccult filariasis and tropical pulmonary eosinophilia

Clinically asymptomatic High eosinophil count

Microfilariae in the tissues

2  Acute disease

3  Chronic disease

K23346_Neligan_15_Filaria_r3_0215-0234.indd 218

5/26/2015 9:09:40 AM

Chapter 15  Filaria

219

FIG. 15-4  This 32-year-old patient from an endemic area has extensive involvement and deformity of the penis and scrotum.

Four grades of lymphedema have been described, depending on the severity of involvement and the quality of pitting.3 • Grade 1: Edema appears and disappears spontaneously and is pitting and uniform in size. • Grade 2: Edema is persistent, pitting, and uniform. • Grade 3: Edema is persistent, nonpitting, and uniform. • Grade 4: Giant lymphedema develops, with complications such as ulcers, warty growth, and loss of limb shape (elephantiasis). Any breach of skin integrity of the affected region (for example, from injury, fungal or bacterial infection, or even eczema) favors entry of pathogenic bacteria into the tissues, leading to acute attacks of adenolymphangitis, which is commonly seen in filarial limbs.4,5 Adenolymphangitis is one of the important clinical manifestations of lymphatic filariasis. Recurrence contributes to the progress of the disease and has important socioeconomic implications, because it affects a patient’s ability to work.6 Lymphedema of the extremities is a common chronic manifestation of lymphatic filariasis that results in elephantiasis as it progresses. Elephantiasis refers to massive swelling of the lower limbs caused by repeat attacks of filarial lymphangitis over several years. The limb becomes grossly enlarged, resembling the foot of an elephant. Repeated inflammatory reactions cause vessel dilation and thickening. In the advanced stages of lymphedema, the skin is thickened and thrown into folds, often with hypertrichosis, black pigmentation, nodules, warty growth, and intertrigo in the webs of the toes, with nonhealing ulcers. This usually involves the lower limbs, commonly unilaterally. The male genitalia are also commonly affected (Fig. 15-4). Vulval elephantiasis has been reported.7

Diagnosis Patient history is essential for diagnosing lymphatic filariasis, because it provides information about the causes. Careful clinical examination of the skin color, texture, and other changes is helpful for staging the disease. Circumferential measurements at fixed points of the upper and lower limbs are documented. The patient’s height and weight are recorded.

K23346_Neligan_15_Filaria_r3_0215-0234.indd 219

5/26/2015 9:09:41 AM

220

Part III  Pathophysiology and Clinical Presentation

Several diagnostic tools are available, including the following: • An immunochromatographic test can be conducted quickly and easily at the bedside to test for lymphatic filariasis. The test is highly sensitive for W. bancrofti. • Ultrasonography can be used as a screening test in endemic areas. It can reveal moving adult worms in the scrotum or breast. Patients with positive results may not have clinical signs and symptoms; removing the adult worms surgically from these patients will prevent the occurrence of lymphatic filariasis. • Lymphoscintigraphy is the single most useful tool in establishing the diagnosis, grading, and cause (see Chapter 26). It can show the outcome of treatment after chemotherapy or surgery. • MRI is useful in the presence of associated problems. MR lymphangiography is a very good assessment modality but very expensive in a country such as India, where health coverage by insurance or the government is not available (see Chapter 28).

Assessment Lymphedema can be objectively assessed by several methods, ranging from clinical evaluation to radiologic modalities. One clinical method involves measuring the limb circumference at various points. The upper limb is divided into four segments. The upper limit of measurement, known as the 65% point, is marked on the upper arm 65% of the distance from the olecranon to the acromion tip. The four segments are (1) the wrist at the level of ulnar styloid to the midforearm, (2) the midforearm to the elbow at the level of olecranon, (3) the elbow to the midarm, and (4) the midarm to the 65% point. These segments conform to the shape of a truncated cone. Measurements obtained using this method produce the least standard error of measurement. The volume of each segment can be calculated using the following formula: Segment volume 5 h(C12 1 C22 1 [C1 3 C2])/12p where h is the length of each segment and C1 and C2 are the circumference of each segment at both ends. The sum of the segment volumes is the volume of the limb.8 Another clinical method is water displacement volumetry. The patient’s upper limb is immersed in a graduated steel cylinder up to the 65% point. The volume of water displaced can be calculated using the following formula: Volume 5 pr2h where r is the radius of the cylinder and h is the height of the water displaced. Calculation of the volume of edema in unilateral cases is recommended. This is done by estimating the difference in the limb volume between the edematous limb and the normal limb. In lymphedema patients, high-frequency sonography is useful for assessing the thickness of the skin and the subcutaneous tissue. The patient can be sitting or lying down, with the limb extended to allow access to all aspects—anterior, posterior, medial, and lateral—to determine the average value. Compressibility of the regional veins can be assessed to rule out venous thrombosis.

K23346_Neligan_15_Filaria_r3_0215-0234.indd 220

5/26/2015 9:09:41 AM

Chapter 15  Filaria

221

Lymphoscintigraphy using technetium sulphur colloid is a useful modality for studying the lymphatics preoperatively and postoperatively. It reliably allows visualization of the lymphatic vessels and lymph flow and is helpful in distinguishing between primary and secondary lymphedema.9 It is performed after intradermal injection of Tc99m (technetium) colloid in the web spaces of the lower limbs (see Chapter 26).

Medical Management The treatment options for patients with lymphedema are broadly divided into two categories: medical and surgical. Many methods used in the past, such as crepe bandaging and flap transfers, are now rarely used. The following recommendations apply to all four grades of lymphedema: • Foot care • Avoidance of injuries and injections to the affected limb • Elimination of the focus of sepsis • Complete decongestive therapy with bandaging, followed by placement of pressure garments Complete decongestive therapy is an important modality in the management of lymphedema. It consists of manual manipulation of the lymphatic ducts, short stretch compression bandaging, therapeutic exercises, and meticulous skin care. • Elevation of the affected part • Management of acute attacks • Cyclic chemotherapy (antibiotic and antifilarial) to prevent secondary infection

Maintenance of Limb Hygiene Filarial patients with damaged lymphatic vessels often have more bacteria on the skin than those without this disease; hence infections are very common. The large number of bacteria on the skin, multiple skin lesions, slow lymph fluid movement, and the reduced ability of the lymph nodes to filter bacteria cause inflammation characteristic of an acute attack. Washing the limb is essential to block this vicious cycle. Clean water at room temperature should be used. The least expensive soap without perfume is usually best. After the area is washed, it should be gently towel dried. This should be done twice daily, ideally in the morning and at night.

Prevention and Treatment of Entry Lesions Apart from the bite sites of the mosquitoes, other skin lesions are common in patients at risk of developing lymphedema and in those who already have it. These are most frequently found between the toes and deep skin folds and around the toenails. Dental caries are another source of secondary infection. The lesions allow bacteria or fungi to enter the body through the skin, and this can cause attacks of acute dermatoadenolymphangitis. Bacterial infections leak fluid that is watery and clear or viscous and colored at times. Fungal lesions are usually white or pink and do not leak fluid. Fluid leakage in bacterial infections is not chylous reflux, which essentially is white. Treatment involves the application of local antibacterial and antifungal creams at least twice daily. Prevention of secondary infection in the initial stages of lymphatic filariasis can reverse or minimize the damage done by the parasite10 (see Chapter 29).

K23346_Neligan_15_Filaria_r3_0215-0234.indd 221

5/26/2015 9:09:41 AM

222

Part III  Pathophysiology and Clinical Presentation

Elevation of the Limb Elevating the limb is important for patients with lymphedema, because it helps to prevent fluid from accumulating in the leg by improving lymph outflow. When the patient is sitting, the foot should be raised to a comfortable height, preferably as high as the hip.

Exercise and Footwear Exercise is useful for patients with lymphedema, because it helps to pump fluid and improve drainage. The more a patient exercises, the better the fluid drainage will be proximally. No exercise should be undertaken during acute attacks. While standing, the patient should perform heel-raising exercises five to ten times or as often as is comfortable. Patients should wear proper footwear and avoid tight-fitting shoes.

Management of Acute Attacks The onset of acute attacks in lymphatic filariasis indicates continuing destruction of the lymphatic system. Any reduction in the frequency of such attacks is an indication that the patient’s condition is improving. Patients should rest and elevate the limb comfortably as much as possible, as indicated previously. A cloth soaked in cool, clean water and placed around the limb can relieve pain. The limb should be gently and carefully washed with soap and clean water. After the skin is dry, an antiseptic should be applied, along with an antibiotic ointment. Patients should drink plenty of water. Paracetamol (acetaminophen) can be taken for fever every 6 hours until the fever lessens. Parenteral antibiotics are recommended during acute attacks, because they can shorten the duration of the attack.

Cyclic Chemotherapy Chemotherapeutic management involves diethylcarbamazine alone, diethylcarbamazine and albendazole, or diethylcarbamazine and ivermectin, along with periodic antibiotics such as penicillin, doxycyline, and sulphonamides. Doxycycline is very useful for treating symbiotic bacterial infections with Wolbachia, which reside inside the parasite and cause resistance to antifilarial drugs (Fig. 15-5) (see Chapter 30).

A

B l u

u

c

c h

u

h u

m

l

FIG. 15-5  Symbiotic infection with Wolbachia. A, Live organisms. B, Dead organisms after treatment with chemotherapy.

K23346_Neligan_15_Filaria_r3_0215-0234.indd 222

5/26/2015 9:09:41 AM

Chapter 15  Filaria

223

Management of Grades III and IV Lymphedema In addition to the basic recommendations described previously, patients with grades III or IV lymphedema ideally should undergo surgical correction.

Surgical Management Various surgical procedures offer relief from lymphedema; however, there is no consensus on surgical intervention or the type and timing of procedures to be performed in a given case. The various surgical approaches to lymphedema treatment are covered in detail in other chapters. Our policy is to combine physiologic surgery such as a nodovenous shunt or a lymphaticovenous shunt with an immediate reduction or debulking procedure without skin grafting. After more than 25 years of experience, we have perfected the technique. The functional and aesthetic aspects of the limb are preserved through microvascular surgery, including free lymphatic channel transfer and lymph node transfer. Omental transfer and supermicrovascular surgery such as lymphaticlymphatic anastomosis are useful for treating congenital and postoperative lymphedema. These strategies have not traditionally played a strong role in the treatment of lymphatic filariasis.

Direct Excision Direct excision involves the removal of skin and subcutaneous tissues circumferentially to the level of the deep fascia; the resultant wound is then covered with a split-skin graft.11 In our experience, this procedure is the most common surgery performed for moderate to advanced stages of chronic lymphatic filariasis of an extremity (see Table 15-1). It results in satisfactory appearances and quick debulking of the lymphedematous tissues in one operation in India (Fig. 15-6) (see Chapter 31).

Scrotoplasty With Penile Debulking and Grafting Total subcutaneous excision followed by split- or full-thickness skin grafts is primarily indicated for primary lymphedema and for secondary lymphedema if the lymphatic trunks are sclerosed and unsuitable for microlymphatic anastomosis or if conservative measures were unsuccessful or not applicable.12 In cases of penile involvement (ram’s horn penis), penile debulking to the level of the tunica albuginea followed by skin grafting provides excellent cosmetic and functional results.13

Agarwal Surgical Technique of Direct Excision The Agarwal technique of direct excision is performed preferably with a regional block with tourniquet control. The involved area is excised down to the level of the deep fascia. The entire skin and subcutaneous tissue containing dilated vessels and lymphedematous tissue is excised. Once excision is complete, hemostasis is secured and confirmed. The wound is then covered by a splitthickness skin graft. In this technique, the deep fascia is preserved, which serves two purposes. First, the fascia facilitates better drainage of both venous blood and lymph, and second, it is a good bed for the split-skin graft.

K23346_Neligan_15_Filaria_r3_0215-0234.indd 223

5/26/2015 9:09:41 AM

224

Part III  Pathophysiology and Clinical Presentation

A

B

C

D

E

F

FIG. 15-6  This 25-year-old woman had chronic lymphatic filariasis (A-C). She is shown 1 year after direct excision and skin grafting on the foot region only. The cosmetic result is further enhanced by her functional ability to wear footwear, although the leg has not been debulked (D-F).

Excision with skin grafting gives immediate and satisfactory long-term results and is now becoming the treatment of choice for moderate to large chronic lymphatic filariasis of the leg and for chronic genital lymphedema.14 Postoperative Care Postoperatively the graft requires regular care and evaluations. The first dressing should be changed after 48 hours to make certain that no hematomas or seromas develop underneath the graft. Thereafter the dressings are changed every 2 to 3 days, because the wounds often produce a lot of exudate and discharge. The graft usually takes well. Patients are then advised to wear pressure garments to prevent hypertrophic scarring. These also help to control edema.

K23346_Neligan_15_Filaria_r3_0215-0234.indd 224

5/26/2015 9:09:44 AM

Chapter 15  Filaria

225

Complications and Treatment Outcomes  The most common complications of this technique of direct excision are graft loss and infection from the high levels of bacteria residing on the skin of these patients. Minimal areas of graft loss are treated conservatively, and the wound is allowed to epithelialize. In patients with large, raw areas, repeat grafting may be needed to decrease the morbidity.

Manokaran Surgical Technique for Debulking The Manokaran surgical technique for debulking is the most useful protocol and provides the best acceptable results. The technique and time duration need to be modified for each patient. All patients are advised to follow the foot hygiene instructions provided, to eliminate the focus of sepsis, to prevent injury to the affected limb, and to preserve the limb shape with the use of pressure garments postoperatively. Stages 3 and 4 of lymphedema cannot be cured, only controlled. The debulking surgical procedure15 in lymphatic filariasis is performed for International Society of Lymphology stage III lymphedemas with nodules, warty growths, and ulcers. The basic principles of surgery are the following: • Augmentation of lymphatic drainage by a physiologic procedure • Reduction of the lymphatic load by debulking the lymphedematous, lymph-producing surface Our strategy involves a procedure to create temporary drainage, such as complete decongestive therapy for 1 week. A permanent drainage procedure is then performed, such as a nodovenous shunt, a lymphaticovenous shunt, a free omental transfer, or a supermicrovascular surgery, transplanting a myocutaneous flap with an arterial, venous, and lymphatic-lymphatic anastomosis. Once permanent lymphatic drainage is established, the grade 4 lymphedema with or without skin changes decreases, leaving only the subcutaneous fat, fibrous tissue, and the soft tissues above the muscle and fascia. We wait 10 to 14 days and then debulk the excess skin, fat, and subcutaneous tissue to the level of the deep fascia, under tourniquet control. This debulking surgery may have to be repeated periodically at a minimum interval of 6 weeks to 3 months, depending on the entire size of the limb, until the shape and size are near normal. We have tried to employ the same skin for resurfacing without using a split-thickness skin graft. The remaining skin with subcutaneous tissues containing the subdermal lymphatics drains the reshaped limb and maintains the contour for a long time. Assistance is provided with a pressure garment, leg elevation, elimination of the focus of sepsis, and prevention of secondary infection by periodic, cyclic antibiotics such as penicillin, doxycycline, and quinolones (ciprofloxacin, ofloxacin, and others), depending on patient sensitivity and the sensitivity pattern of the drug. The outcome of debulking surgery depends on rigidly sequenced preoperative preparation and postoperative follow-up, with the recommendations discussed previously.16 If a patient does not follow the postoperative instructions meticulously, secondary infection often develops. Secondary infection leading to lymphangitis and cellulitis is the main cause for the recurrence and progression of lymphedema. We have used this technique for 25 years with very good results, including maintenance of the shape and size of the limb in our long-term follow-ups. If a patient has recurrence or progression of lymphedema, a lymphoscintigram is performed to determine the status of the lymphatics, lymph nodes, and drainage. Most cases of recurrence are lymphangitis from

K23346_Neligan_15_Filaria_r3_0215-0234.indd 225

5/26/2015 9:09:44 AM

226

Part III  Pathophysiology and Clinical Presentation

noncompliance. We encourage these patients to meticulously follow the conservative, nonsurgical methods such as manual lymph drainage and complete decongestive therapy. Most patients improve with this therapy, and their original limb shape and size return. This is maintained with a pressure garment or bandages. Very few patients (5% to 6%) need a revision surgical procedure, for example, a repeat nodovenous shunt or a lymphaticovenous shunt. This debulking procedure is always done under tourniquet control to prevent blood loss, hematoma, and infection. The tourniquet can be used for up to 2 hours in the lower limb and 1 hour in the upper limb. Once the excision is completed, the tourniquet is released and hemostasis confirmed. The suction drain is left in place, and the wound is closed in layers.17 The incision is always made as a reverse hockey stick shape, on the medial side of the limb. The edges of the skin surface are confirmed to have good viability after excess skin is trimmed. We always try to use the same scar for subsequent reduction surgeries. The excision always stops short of the deep fascia. We never open the deep fascia, because it allows the muscle to bulge into the subcutaneous plane and complicates wound closure. It causes pain postoperatively and can block the suction drains. Complications  Very few complications occur with this surgery. They include the following: • Lymphorrhea • Lymphocele • Wound dehiscence • Hemorrhage Although several morbidities are possible, mortality has not been reported from these surgical procedures. A good preoperative assessment and preparation helps to prevent most unwanted complications postoperatively. In many of the centers where debulking surgery is performed for lymphedema, it is always undertaken as a secondary procedure, after a lymphatic drainage procedure is performed, as described previously.

Other Debulking Procedures Charles’s excision is another debulking procedure that has been performed for a long time (see Chapter 31). The lymphedematous tissue (skin and subcutaneous tissue, including the deep fascia) is excised circumferentially, and a split-thickness skin graft is used to cover the raw area. Because no subdermal plexus is available for drainage and the split-thickness skin graft adheres to the muscle, it produces much worse edema distal to the excision, usually in the foot. This procedure is not commonly performed today because of the bottleneck deformity and unaesthetic outcome. Kondolean’s excision is technically similar to the Charles procedure and is not often performed.16,18 Thompson’s procedure was claimed to be a physiologic procedure, because the deepithelialized dermal flap is buried under the opposite skin flap and sutured in two layers.19 The disadvantage of this procedure is that if the dermal flap is sutured as a deeper layer necroses, then the skin closure will not heal. Therefore the flaps need to be reopened and the necrosed skin flap salvaged. Skin coverage is then provided. This causes morbidity to the affected limb and prolongs healing time. Currently a multiple-stage process consisting of a microvascular lymphatic drainage procedure, a simple elliptical excision, and maintenance by conservative multimodality therapies such as periodic antibiotics, regular foot hygiene, complete decongestive therapy, and pressure garments

K23346_Neligan_15_Filaria_r3_0215-0234.indd 226

5/26/2015 9:09:44 AM

Chapter 15  Filaria

227

provides the most acceptable long-term results. The newer techniques and microvascular surgery play a crucial role in management for more acceptable functional and cosmetic results.

Lymphosuction A number of studies have evaluated the use of liposuction for breast cancer–related upper extremity lymphedema. Brorson and Svensson20 prospectively followed patients and reported overall favorable results. The authors have, however, reported on the use of lymphosuction in the treatment of chronic lymphatic filariasis of long duration. Lymphosuction has been found to be a useful treatment modality for mild to moderate grade II lymphedema, with satisfactory results21 (see Chapter 32).

Flap Interposition In 1950 Gillies22 described the first flap interposition for treatment of lower extremity lymphedema through a two-stage operation in which a flap of skin and subcutaneous tissue was transferred from the arm to the affected groin. This provided a path for lymphatic fluid to bypass the damaged lymphatics in the groin. The procedure, however, was associated with a high incidence of complications when carried out by other surgical groups; therefore it is rarely performed today.

Free Muscle and Myocutaneous Flaps The free latissimus dorsi island myocutaneous flap has been used extensively for reconstruction after mastectomy. The bilateral transverse rectus abdominis myocutaneous flap is also used as free flap for the treatment of upper limb lymphedema. In this era of supermicrovascular surgery, surgeons in Spain and Belgium have tried anastomosing the artery, vein, and the lymphatics at the same time as the mastectomy, followed by reconstruction. They have proved that this can prevent postmastectomy lymphedema. Gracilis muscle and myocutaneous flaps have also been used in similar techniques to prevent lower limb lymphedema.23

Lymph Node Transfer In lymph node transfers, healthy lymph nodes are harvested and transplanted either to the original site of injury or to nonanatomic areas within the lymphedematous limb. The transplantation is performed by simply mincing the lymph nodes and delivering them to the site as an avascular graft or by transferring a vascularized tissue in which the lymph nodes and surrounding fat are intact, repairing their arterial and venous blood supply microsurgically.24 However, the risk of lymphedema in the donor extremity is a concern. Therefore such transfers have not been widely accepted (see Chapter 36)

Lymphaticovenous Bypass Procedures have been described to drain obstructed lymphatic vessels into the venous circulation by surgically creating lymphaticovenous shunts. Initially, these shunts were created using large superficial veins as the outlet vessels; however, venous hypertension with resultant decreased lymphatic outflow led to the use of subdermal venules.25 In India use of this technique is limited, because individuals with lymphatic filariasis typically present at a late stage.

K23346_Neligan_15_Filaria_r3_0215-0234.indd 227

5/26/2015 9:09:44 AM

228

Part III  Pathophysiology and Clinical Presentation

Nodovenous Shunt In 1963 Nielubowicz et al26 developed nodovenous anastomosis27-29 in lymphedema produced artificially in dogs. The procedure was carried out in various parts of world in humans with lymphedema, with positive outcomes. It has become a surgery of choice in early lymphedema in some centers and in patients with elephantiasis and is performed before cytoreduction or debulking procedures. Nodovenous anastomosis is a more physiologic procedure and is very useful in patients with a deformity or disease of the afferent lymphatics (for example, lymphatic filariasis, posttraumatic lymphedema, and postinflammatory lymphedema).30 However, it is not useful in the absence of lymphatics and lymph nodes (for example, after mastectomy or radiotherapy or for congenital lymphedema). Thus the procedure is not very popular in Europe, even though it was first developed there.

Indications Indications for a nodovenous shunt include the following: • Patients with a competent saphenofemoral junction • Patients without inguinal abscess or sepsis • All grades of lymphedema (according to the International Society of Lymphology stages) • A healthy and functioning lymph node or lymph nodes (determined with lymphoscintigraphy or ultrasonography)

Contraindications Contraindications to nodovenous shunt procedures include the following: • No visible lymph node on lymphoscintigraphy or ultrasonography • Associated varicose veins or saphenofemoral incompetence • No reduction of circumferential measurements of the leg at any given point, even after 6 days of manual lymph drainage • Acute adenolymphangitis • Elderly and debilitated patients • Associated medical comorbidities Two methods of anastomosis have been described: end-to-end and end-to-side.

End-to-End Anastomosis A nodovenous shunt for lower limb lymphedema is performed with the patient under general or regional anesthesia and placed in a supine position. A vertical 3 cm incision is made in the upper part of the thigh, just medial to the femoral pulsations, and the long saphenous vein (or another good-caliber vein) is exposed. The distal end is ligated with chromic catgut, and the upper end is opened in the shape of a fish mouth. The proximal segment should have no retrograde flow, confirming that saphenofemoral incompetence is not present. A vertical group of inguinal lymph nodes is identified. These nodes must be pink and at least 1 cm in diameter.31 No dissection is performed around the lymph nodes to preserve afferent and efferent lymphatics. The upper capsule of the lymph nodes is shaved. Lymph will ooze from the cut surface. Diathermy should not be used. Bipolar diathermy causes less damage and can be used if the area needs to be sealed.

K23346_Neligan_15_Filaria_r3_0215-0234.indd 228

5/26/2015 9:09:44 AM

229

Chapter 15  Filaria

A

B

FIG. 15-7  Nodovenous bypass. A, Endto-end. B, End-to-side.

The proximal, cut end of the long saphenous vein is anastomosed to the cut surface of capsule of the node using 6-0 or 7-0 nylon continuous sutures (Fig. 15-7). Hemostasis is confirmed, and the wound is closed in layers. No drain is placed.

End-to-Side Anastomosis Alternatively, a nodovenous shunt can be performed in end-to-side fashion. Vascular clamps are applied to the vein both proximally and distally. A No. 11 blade is used to make a vertical stab incision of 0.5 to 1 cm, depending on the vein caliber, in the surface adjoining the lymph node. The cut surface of the node is anastomosed with the vertical stab incision of the vein using 8-0 nylon interrupted sutures.28,32-34 Clamps are released, and the vein is observed for filling. The anastomosis site is continuously irrigated with heparinized saline to prevent clot formation, which is common in this technique. If a healthy or adequate-sized lymph node cannot be identified in the inguinal region, multiple lymphatic channels can be buried along the length of the vein at three or four sites. Stab incisions are made in the vessel using an 18-gauge needle. The open end of the lymphatics are left to float in the venous lumen. The lymphatic vessels are anchored with 8-0 nylon single, interrupted sutures.

Complications Complications of nodovenous shunts include the following: • Seroma • Lymphorrhea • Lymphocele • Wound dehiscence

K23346_Neligan_15_Filaria_r3_0215-0234.indd 229

5/26/2015 9:09:45 AM

230

Part III  Pathophysiology and Clinical Presentation

Lymphatic-Lymphatic Anastomosis Lymphatic-lymphatic anastomosis is commonly performed to bridge the anatomic discontinuity in patients with congenital lymphedema. This method is useful for lymphedema that develops after a mastectomy. The lymphatics are harvested from the normal lower limb thigh segments. Multiple lymphatic channels are anastomosed with the upper and lower end of the normal lymphatics to bridge the absent lymphatics. An operating microscope with 123 to 143 magnification is used, as with other microsurgical procedures.35-39 We prefer to use 10-0 nylon suture.

Free Omental Transfer Free omental transfer is useful for lymphatic filariasis40 and posttraumatic and postoperative lymphedema. In lymphatic filariasis with lower limb lymphedema, a vertical upper thigh midline incision is made. The long saphenous vein, the superficial circumflex iliac artery, and the inguinal lymph nodes or lymphatics are exposed and prepared for microvascular anastomosis. The abdomen is opened with a lower transverse incision. The omentum is dissected with its artery, vein, and lymphatics, which can be anastomosed with the respective vessels. During the closure of the thigh incision, a small window is left open for assessing the viability of the omentum. The window can be closed secondarily. Hemostasis is confirmed, and the abdomen is closed in layers. The tunneling of omentum into the inguinal region (omentoplasty)41 was initially popular with Russian surgeons for management of various types of lymphedema. Because the continuity of the omentum was maintained, the incidence of lymphangitis of the leg increased, leading to peritonitis. The technique was subsequently abandoned. Surgery for lymphedema should not cause mortality. A certain percentage of morbidity is acceptable if patients’ overall condition improves.

Conclusion Intensive, well-planned, and sequenced conservative treatment of outpatients with all grades of lymphedema can produce significant reduction in the volume of the edema over a short time, even in the most advanced stages of the disease. The frequency of attacks of episodic adenolymphangitis is not related to the administration of specific antibiotics or diethylcarbamazine; for the most part, it is controlled by simple hygienic measures combined with good limb and foot care and the application of a local antibiotic or antifungal cream. If performed appropriately, these measures are effective in reducing the number of acute dermatoadenolymphangitis attacks. Surgical treatment comprising direct excision with skin grafting offers the best hope for patients who are refractory to conservative medical management. Secondary lymphedema is common in developing countries because of filarial worm infestations. Lymphatic filariasis, though not curable completely, can be treated. Morbidity can be managed and aesthetic results obtained. Future treatment in large part will depend on current and upcoming developments in surgical techniques.

K23346_Neligan_15_Filaria_r3_0215-0234.indd 230

5/26/2015 9:09:45 AM

Chapter 15  Filaria

231

C linical P earls • Early, aggressive treatment in chronic lymphatic filariasis is essential for satisfactory results. • Lymphoscintigraphy is the most helpful modality for establishing a diagnosis and for assessing the effectiveness of treatments. • Cases of recurrence after debulking are typically lymphangitis from noncompliance, most of which are effectively treated with meticulous nonsurgical methods. • Deep fascia should not be opened in debulking procedures. • Surgical treatment comprising direct excision with skin grafting offers the best hope for patients who are refractory to conservative medical management. • Nodovenous shunt procedures are most useful in patients with diseases of the afferent lymphatics but are not effective in the absence of lymphatics and lymph nodes.

ACKNOWLEDGMENT Dr. Manokaran would like to thank all of his patients who made this study possible. He would also like to thank Drs. V. Rajmohan and K. Haripriya (research scholar), who have contributed to his work at various times. Drs. Agarwal would like to gratefully acknowledge the help of Professor Ramesh Chandra in proofreading the manuscript and Professor Padam K. Agarwal in helping with the histopathologic interpretation of the tissue specimens.

R EFERENCES 1. Shenoy RK. Clinical and pathological aspects of filarial lymphedema and its management. Korean J Parasitol 46:119-125, 2008. 2. Pani SP, Yuvaraj J, Vanamail P, et al. Episodic adenolymphangitis and lymphedema in patients with bancroftian filariasis. Trans Roy Soc Trop Med Hyg 89:72-74, 1995. 3. Kumaraswami V. The clinical manifestations of lymphatic filariasis. In Nutman TB, ed. Lymphatic Filariasis. London: Imperial College Press, 2000. 4. Shenoy RK, Kumaraswami V, Suma TK, et al. A double-blind, placebo-controlled study of the efficacy of oral penicillin, diethylcarbamazine or local treatment of the affected limb in preventing acute adenolymphangitis in lymphedema caused by brugian filariasis. Ann Trop Med Parasitol 93:367-377, 1999. 5. Suma TK, Shenoy RK, Varghese J, et al. Estimation of ASO titer as an indicator of streptococcal infection precipitating acute adenolymphangitis in brugian lymphatic filariasis. Southeast Asian J Trop Med Pub Health 28:826-830, 1997. 6. Shenoy RK, Sandhya K, Suma TK, et al. A preliminary study of filariasis related acute adenolymphangitis with special reference to precipitating factors and treatment modalities. Southeast Asian J Trop Med Public Health 26:301-305, 1995.

K23346_Neligan_15_Filaria_r3_0215-0234.indd 231

5/26/2015 9:09:45 AM

232

Part III  Pathophysiology and Clinical Presentation

7. Khanna NN, Joshi GK. Elephantiasis of female genitalia: case report. Plast Reconstr Surg 48:379-381, 1971. 8. Taylor R, Jayasinghe UW, Koelmeyer L, et al. Reliability and validity of arm volume measurements for assessment for lymphedema. Phys Ther 86:205-214, 2006. 9. Yuan Z, Chen L, Luo Q, et al. The role of radionuclide lymphoscintigraphy in extremity lymphedema. Ann Nucl Med 20:341-344, 2006. 10. Calnan JS. New concepts of the function of the lymphatic system in the swollen leg. Sci Basis Med Annu Rev 349-364, 1971. 11. Thompson N. The surgical treatment of chronic lymphoedema of the extremities. Surg Clin North Am 47:445-503, 1967. 12. Manokaran G. Management of genital manifestations of lymphatic filariasis. Ind J Urol 21:39-43, 2005. 13. Modolin M, Mitre AI, da Silva JC, et al. Surgical treatment of lymphedema of the penis and scrotum. Clinics (São Paulo) 61:289-294, 2006. 14. Morey AF, Meng MV, McAninch JW. Skin graft reconstruction of chronic genital lymphedema. Urology 50:423-426, 1997. 15. Handley WS. Hunterian lectures on the surgery of the lymphatic system. Br Med J 1:922-928, 1910. 16. Kondoleon E. Die operative Behandlung der elephantiastichen Oedeme. Zentralbl Chir 39:1022, 1912. 17. Larson DL, Coers CR, Doyle JE, et al. Lymphedema of the lower extremity. Plast Reconstr Surg 38:293301, 1966. 18. Servelle M. Surgical treatment of lymphoedema: a report on 652 cases. Surgery 101:485-495, 1987. 19. Sawhney CP. Evaluation of Thompson’s buried dermal flap operation for lymphoedema of the limbs: a clinical and radioisotopic study. Br J Plast Surg 27:278-283, 1974. 20. Brorson H, Svensson H. Liposuction combined with controlled compression therapy reduces arm lymphedema more effectively than controlled compression therapy alone. Plast Reconstr Surg 102:10581067; discussion 1068, 1998. 21. Agarwal R, Bhatnagar SK, Chandra R. Lymphosuction: a new treatment modality for chronic filarial lymphedema. Eur J Plast Surg 21:113-117, 1998. 22. Gillies H. The lymphatic wick. Proc R Soc Med 43:1054-1056, 1950. 23. Handley WS. Lymphangioplasty: a new method for the relief of the brawny edema of breast cancer and for similar conditions of lymphatic oedema: preliminary note. Lancet 1:783-785, 1908. 24. Lin CH, Ali R, Chen SC, et al. Vascularized groin lymph node transfer using the wrist as a recipient site for management of postmastectomy upper extremity lymphedema. Plast Reconstr Surg 123:12651275, 2009. 25. Huang GK, Hu RQ, Liu ZZ, et al. Microlymphaticovenous anastomosis in the treatment of lower limb obstructive lymphedema. Analysis of 91 cases. Plast Reconstr Surg 76:671-685, 1985. 26. Nielubowicz J, Olszewski W, Sokolowski J. Surgical lympho-venous shunts. J Cardiovasc Surg 9:262267, 1986. 27. Gloviczki PJ, Hollier LH, Nora FE, et al. The natural history of microsurgical lymphovenous anastomoses: an experimental study. J Vasc Surg 4:148-156, 1986. 28. Gloviczki PJ, Fisher J, Hollier LH, et al. Microsurgical lymphovenous anastomosis for treatment of lymphedema: a critical review. J Vasc Surg 7:647-652, 1988. 29. Olszewski WL. The treatment of lymphedema of the extremities with microsurgical lympho-venous anastomoses. Int Angiol 7:312-321, 1988. 30. Damstra RJ, Voesten HG, van Schelven WD, et al. Lymphatic venous anastomosis (LVA) for treatment of secondary arm lymphedema. A prospective study of 11 LVA procedures in 10 patients with breast cancer related lymphedema and a critical review of the literature. Breast Cancer Res Treat 113:199-206, 2009. 31. O’Brien BM, Mellow CG, Khazanchi RK, et al. Long-term results after microlymphaticovenous anastomoses for the treatment of obstructive lymphedema. Plast Reconstr Surg 85:562-572, 1990.

K23346_Neligan_15_Filaria_r3_0215-0234.indd 232

5/26/2015 9:09:45 AM

Chapter 15  Filaria

233

32. Huang GK. [Results of microsurgical lymphovenous anastomoses in lymphedema: report of 110 cases] Langenbecks Arch Chir 374:194-199, 1989. 33. Yamamoto Y, Sugihara T. Microsurgical lymphaticovenous implantation for the treatment of chronic lymphedema. Plast Reconstr Surg 101:157-161, 1998. 34. Zolotorevskiĭ Vla, Savchenko TV, Chernysheva LM, et al. [Late results of lymphovenous anastomoses in lymphedema of the lower extremities] Khirurglia (Mosk) 5:96-101, 1990. 35. Campisi C, Boccardo F. Lymphedema and microsurgery. Microsurgery 22:74-80, 2002. 36. Campisi C, Boccardo F. Microsurgical techniques for lymphedema treatment: derivative lymphaticvenous microsurgery. World J Surg 28:609-613, 2004. 37. Campisi C, Davini D, Bellini C, et al. Lymphatic microsurgery for the treatment of lymphedema. Microsurgery 26:65-69, 2006. 38. Campisi C, Eretta C, Pertile D, et al. Microsurgery for treatment of peripheral lymphedema: long-term outcome and future perspectives. Microsurgery 27:333-338, 2007. 39. Clodius L, Piller NB, Casley-Smith JR. The problems of lymphatic microsurgery for lymphedema. Lymphology 14:69-76, 1981. 40. Goldsmith HS, De los Santos R, Beattie EJ Jr. Relief of chronic lymphedema by omental transposition. Ann Surg 166:572-585, 1967. 41. Silver D, Puckett CL. Lymphangioplasty: a ten year evaluation. Surgery 80:748-755, 1976.

K23346_Neligan_15_Filaria_r3_0215-0234.indd 233

5/26/2015 9:09:45 AM

K23346_Neligan_15_Filaria_r3_0215-0234.indd 234

5/26/2015 9:09:45 AM

C hapter 16 Lymphedema in Pediatric Patients Cristobal Miguel Papendieck

K ey P oints • Pediatric lymphedema is a chronic, impairing, progressive disorder. • Each age group within the pediatric range has its specific and distinguishing issues and needs. • Comprehensive, multifaceted holistic treatment and management produce the best outcomes. • A constant in the treatment of primary and secondary lymphedemas in pediatric patients is appropriate and adequate vascular rehabilitation. • If a surgical option is considered, the most common one is lymphovenous anastomosis.

Lym Lymphedema, regardless of form and location, is a condition that has a low profile in the literature despite its prevalence; it is an orphan in the medical and pediatric world, even though one third of the lymphedema in the world affects pediatric populations (0 to 14 years).1-9 It is important to note that a primary lymphedema patient may have an expectation of 80-year survival, whereas survival for individuals with secondary lymphedema is estimated to be only approximately 20 years. This means a longer burden on the patient, the family, the health care system, and society if the lymphedema is not recognized, treated, and managed appropriately in the very early stages. Furthermore, serious life-threatening side effects of chronic lymphedema, such as Stewart-Treves syndrome,9 may occur more frequently in the primary lymphedema group because of its duration and severity, but there are no statistics regarding this. Primary lymphedema is expressed in pediatric patients as congenital, although it is not always known or identified at birth because of a lack of awareness of its possibility or a lack of screening for it.8 One or both lower limbs are the most frequently involved initially (Fig. 16-1), but the condition may present in any part of the body. It is thought to be the result of underdevelopment of the superficial lymphatic system, which results in the presence of abnormally high levels of proteins and tissue fluids in the interstitial space. Exploration of the underlying genetic causes of certain types of primary lymphedema is helping researchers identify and understand previously unrecognized syndromes. 235

K23346_Neligan_16_Lymph in Ped_r3_dc_0235-0246.indd 235

5/26/2015 9:11:06 AM

236

Part III  Pathophysiology and Clinical Presentation

FIG. 16-1  Primary lymphedema in the foot of a newborn. The Stemmer sign in this infant was positive and the pitting test negative.

For primary lymphedemas (as well as secondary ones), distribution around the world is varied not only in terms of incidence and cause but also in the availability of the means for early and accurate diagnosis (sometimes lymphedema is not recognized until a vigilant therapist is consulted), the priority it is given, individual sociocultural status, and access to specialist treatment and management centers.

Diagnosis of Lymphedema in Pediatric Patients Lymphedema (a swelling of the affected area) in pediatric patients is a sign—not a symptom. It is a sign of a disorder or malformation of some section of the lymphatic system.10 It indicates the lymphatic system’s inability to evacuate and transport its specific fluid load (and its contents), which a healthy lymphatic system is supposed to return from the interstitial spaces to the truncal venous system. The interstitial space is where lymph is produced and where all solutes originate that will eventually drain into the venous system. Lymph is an indispensable necessity for vertebrate life.11 Any incompetence of the venous system for carrying its volume generates a potential overload in the lymphatic system, which is known as phlebolymphedema. The regulating mechanism of lymph formation is an activity that obviously never equals zero: lymph is “lymph” only when it enters the lymphatic system precapillaries; before that it is interstitial fluid with two pathways available for its removal, the lymphatics and the venous system. All lymphedemas have interstitial but not necessarily lymphatic hypertension. Lymphatic hypertension is common in secondary noninflammatory lymphedemas, but not in primary lymphedemas. This knowledge is very important when making therapeutic decisions, because a surgically created lymphovenous shunt works best if there is no pressure gradient between the venous and lymphatic systems. The part of the lymphatic system that is very important but which we do not talk about very often is that which transports chyle between the jejunum-ileum and the Pecquet cistern, and then as mixed lymph to the exit of the thoracic duct at the left shoulder (Fig. 16-2).

K23346_Neligan_16_Lymph in Ped_r3_dc_0235-0246.indd 236

5/26/2015 9:11:06 AM

Chapter 16  Lymphedema in Pediatric Patients

237

FIG. 16-2  Chylous reflux (chyle and systemic lymph) seen during surgery on a primary lymphedema.

TABLE 16-1   Causes of Primary Lymphedema Cause

Description

Interstitial

Malfunctioning/dysplasia/hypoplasia of the interstitial lymphatic “endothelial” segment. As with all lymphedemas, this subgroup has only interstitial hypertension and no vessel hypertension or overload because lymph cannot be formed; substances normally transported in the lymph cannot enter the circuit. Essentially, this situation depends on the vascular endothelial growth factors and receptors related to this endothelial segment. Three well-known and identified mutations fall within this group: • VEGFR-3 Milroy disease14 OMIM 154100 • FOXC2 Lymphedema-distichiasis syndrome15,16 • SOX18 Hypotrichosis-lymphedema-telangiectasia syndrome OMIM 60782317

Canalicular (lymphatic vessel)

Lymphangiodysplasias (LAD I): dysplasia of the lymphatic vessels

Nodal

Lymphadenodysplasias (LAD II): dysplasia of the lymph nodes

Eventually both forms of lymphangiodysplasias above together lead to LAAD, lymphangioadenodysplasias.

Primary and Secondary Lymphedema in Pediatric Patients Primary Lymphedema Primary lymphedema is any lymphedema associated with an intrinsic or primary malfunction of the lymphatic system in all or one of the three hemicircuits, as described in Table 16-1. This may be because of interstitial issues such as fibrosis, malformations of the lymphatic vessels or nodes.12 Interestingly it now seems likely that many apparently secondary lymphedemas may have an underlying primary component.13 This is something we need to consider in the future in terms of early identification and anticipation of such problems (Fig. 16-3).

K23346_Neligan_16_Lymph in Ped_r3_dc_0235-0246.indd 237

5/26/2015 9:11:07 AM

238

Part III  Pathophysiology and Clinical Presentation

FIG. 16-3  Primary lymphedema on the upper limb of a newborn. Stemmer sign positive, pitting test negative.

Hypoplasia of the lymphatic system, or hyperplasia or agenesis, either canalicular (lymph vessel) or of the lymph nodes (nodal), is not correctly called dysplasia unless it involves a short or regional system segment. In our experience, hypoplasias or hyperplasias occur more frequently than indicated in the literature about primary lymphedema. The same is true for hypotrophy. What is important to note is that the number and/or size of vessels or lymph nodes does not guarantee functionality or functional quality. Many agenesis-induced lymphedemas actually have hyperplasia of many lymph vessels. Not all dysplasias are enough to account for primary lymphedema. What is more, some dysplasias are never associated with lymphedema per se; for example, macrocystic lymphangiomas and cystic hygromas. Thus lymphedema in pediatric patients is a chronic, impairing, progressive disorder. This chronicity has, as also occurs in secondary lymphedemas, a regional lipogenic effect.18,19 We have much to learn and many problems to solve in terms of our knowledge of the underlying issues of the formation of the lymphatic system and of the treatment of it, especially in pediatric cases.

Syndromes Associated With Primary Lymphedema There are approximately 144 syndromes associated with primary lymphedema (Fig. 16-4).20 Among them, 22 are inherited with lymphatic malformations. Box 16-1 lists some of these conditions.21-44 In addition, primary lymphedemas can be caused by malformation (LAD I and II), with or without chylous reflux, and can occur in combined angiodysplastic syndromes, as follows: • Klippel-Trenaunay-Weber syndrome: Nevus, port-wine stain, varicose veins, osteohypertrophy, overgrowth, micro AV shunts45 • Klippel-Trenaunay-Servelle syndrome: Port-wine stain, varicose veins, osteohypertrophy, overgrowth, truncular venous malformation46,47 • F.P. Weber syndrome: Port-wine stain, macro AV shunt, osteohypertrophy, overgrowth48

K23346_Neligan_16_Lymph in Ped_r3_dc_0235-0246.indd 238

5/26/2015 9:11:07 AM

Chapter 16  Lymphedema in Pediatric Patients

239

• Proteus syndrome: Tridermal and trisystemic vascular anomalies, overgrowth49,50 • CLOVES: Congenital lipomatous overgrowth, vascular malformation, epidermal nevi, primary or secondary scoliosis51 • N1 and N2 syndromes, including Von Recklinghausen syndrome: Neurofibromatosis, Gorrham-Stout-Haferkamp syndrome,52-54 intraosseous lymphangioma

A B

FIG. 16-4  Pediatric patients with primary lymphedema with A, Kasabach Merritt syndrome, and B, neurofibromatosis.

BOX 16-1  Syndromes That Can Occur With Primary Lymphedema Yellow nail syndrome Opitz syndrome Noonan syndrome Turner syndrome PEHO syndrome Aplasia cutis Bronspiegel syndrome Carbohydrate-deficient glycoprotein syndrome Cerebellar hypoplasia Lissencephaly Choanal atresia Cholestasis Aagenaes syndrome Lymphangiectasia Skeletal dysplasia Wässer syndrome Persistent Müllerian duct syndrome Polydactyly

K23346_Neligan_16_Lymph in Ped_r3_dc_0235-0246.indd 239

Cerebral AV malformations Avasthey syndrome Cleft palate Figueroa syndrome Hypoparathyroidism Dahlberg syndrome Leukemia-deafness Emberger syndrome Microencephaly Chorioretinopathy Microcephaly Cutis gyrata Mental retardation Müke syndrome Facial dysmorphia Aortic coarctation Nevo syndrome Hennekam syndrome

5/26/2015 9:11:07 AM

240

Part III  Pathophysiology and Clinical Presentation

Secondary Lymphedema Secondary lymphedema in pediatric patients can occur from a range of causes and situations, including the following: 1. Parasitic lymphedemas (filariasis). The most frequent and aggressive filaria that causes lymphedema is Wuchereria bancrofti. This worm may be as long as 11 cm and may block the lymph node system in any part of the body, but the nodes most affected are those draining the area related to the biting preferences of the mosquito. The host is human; the vector is the female Anopheles mosquito that inoculates microfilaria through its bite and capillary aspiration. Microfilaria larvae are deposited on the skin of the next carrier 24 to 96 hours after the parasite’s metamorphosis. The population at risk is about 1200 million inhabitants, mainly on the Equatorial belt, 30% of whom are children in the 0 to 14 age group. The infected population is more than 120 million, with an estimated 70 million with clinically manifest lymphedema.55 2. Podoconiosis.56,57 This is a regional disease in tropical volcanic farming areas at elevations lower than 1200 meters (because of the temperature and the conditions of work of the farmers who are most often limited to this area). The most affected country is Ethiopia.58 Podoconiosis involves the lower limbs as a result of the plantar absorption of volcanic minerals, which are often rich in silica. Most often it involves a nodal block at the groin.59 3. Phlebolymphedema. This lymphedema results from a truncular venous malformation60,61 with chronic venous hypertension. It is frequently associated with congenital lymphangioadenodyplasia. This is possibly the most frequent nonparasitic secondary lymphedema presentation in pediatric patients. It may be observed in big angiodysplastic syndromes (BAS)62 combined, such as Klippel-Trenaunay-Weber63,64 Servelle,65 Proteo,66,67 and CLOVES68 syndromes and others. 4. Trauma-induced lymphedemas. • Direct trauma, generally soft tissue related (but that later form scars) within and around the lymphatic/vascular systems. • Indirect trauma, inadequate vascular access, radiotherapy, surgery. • Infections, mainly specific and chronic infections such as tuberculosis (Koch bacillus), Calmette-Guerin bacillus, and brucellosis or Malta fever (Brucella melitensis). 5. “Puffy hand” caused by frequent vascular access associated with drug abuse and overuse, which most frequently occurs at the left elbow fold. 6. Side effects and the interaction of medications (chronic anticonvulsant therapy, hormonal medications). 7. Hair tourniquet syndrome.69 8. Amniotic band syndrome.70 9. Collagen disorders.

Treatment of Lymphedema in Pediatrics A constant in the treatment of primary and secondary lymphedemas in pediatrics is adequate vascular rehabilitation. To this end, we consider it essential to follow the clearly established guidelines of the international consensus documents of the International Society of Lymphology (ISL), the International Lymphedema Framework (ILF), and the Latin American Consensus about Lymphedema (LA). We use Vodder’s technique and follow Földi’s method. For bandaging, we use the

K23346_Neligan_16_Lymph in Ped_r3_dc_0235-0246.indd 240

5/26/2015 9:11:07 AM

Chapter 16  Lymphedema in Pediatric Patients

241

multilayer system, with tailor-made soft compression nonelastic or low elastic supports. Skin care is an essential aspect of holistic care for these children. Sequential pumps are not generally used because of the small size of patients’ limbs and relatively large compartment size of the pumps, and because the pressures applied by these devices may be too high given the small circumference of the limbs in young patients.

General Considerations Care of pediatric patients is different and often more complex than that of adult patients. Newborns, infants, young children, and those approaching puberty each have their specific and distinguishing issues and needs.71,72 Unfortunately, relatively little attention has been focused in the literature on the specific care requirements of these children. Pediatric patients require more time, a dedicated health care space, specific psychophysical considerations, and a regard for the complex social aspects of lymphedema in a young person. We use the Porot test73 to determine the patient’s family inclusion, and the free drawing test to interpret psychosocial commitment. We take into consideration other relevant social aspects such as employment, schooling, distances from care facilities, the necessary parental commitment, costs, and the time factor associated with parental involvement. It is important to consider that the mother and father are often young when they suddenly face the diagnosis of such an unexpected and significant disorder in their child. The ongoing nature of treatment for these children can compromise the couple’s relationship and stability, leading to social and relationship issues. It may be useful to encourage involvement in pediatric patient group meetings through which patients and families are brought together who have a similar evolution and similar social and cultural status, the latter being particularly important. When secondary lymphedemas occur, they require specific medical and surgical therapies. For instance, to prevent erysipelas, we always prescribe antibiotics for 1 year with follow-up, to be repeated every 3 years. All patients with lymphedema with lymphatic vessel hypertension in some or all segments may benefit from shunt surgery. This is recommended for pediatric patients with primary lymphedema if their lymphatic hypertension is of the nature described earlier. Thus primary “interstitial” lymphedemas are excluded, but shunt surgery can be beneficial in secondary lymphedema. Similarly, all patients with lymphedema with chylous reflux74-76 to the lower limbs benefit from this technique. Generally, the greater the reflux, the better the outcome. In terms of their role in pediatric secondary lymphedemas, each of these proposed techniques can be considered when appropriate, but their application does not depend solely on one’s training. These are procedures for a lifetime, which means that the significance of the other adjuvant therapies and what they can achieve in remediation of the condition may provide a guide for when they might be used as a supplement to them. As an example, liposuction of fatty tissue in secondary lymphedema,77 which requires elastic support for life, is a questionable approach in pediatric patients. Similarly, the use of techniques requiring local and prolonged anesthesia is not possible, particularly in the younger pediatric group; VEGF-R is not approved for pediatric patients. Terminoterminal or terminolateral anastomosis between lymph vessels or lymphatic venous anastomosis between a normal sectioned vessel is not the same as between anarchic or malformed vessels. Moreover, the caliber of children’s vessels is different, often smaller, than that of adults. We may

K23346_Neligan_16_Lymph in Ped_r3_dc_0235-0246.indd 241

5/26/2015 9:11:07 AM

242

Part III  Pathophysiology and Clinical Presentation

often consider some form of vascular rehabilitation, but this cannot be the sole indication or care plan for the projected 80 years of a young patient’s life.

Surgery in Pediatric Cases In our experience, the most frequent surgical technique used is that proposed by Olszewski.78 but we do not exclude other techniques, such as those proposed by Campisi and colleagues.79-81 We use lymph node venous anastomosis, similar to that of Olszewski, for several reasons: it is not microsurgery, an anastomosis means the involvement in one step of several vessels, and in general, multiplicity of this anastomosis is generally possible. Other issues that should be taken into account include the difficulty of identifying malformed lymph nodes, the significance of the risk of inadequate healing, and venous hypertension, which is a frequent factor in combined angiodysplastic syndromes. Surgery is useful only if lymph nodes are not mobilized, anastomosis is lateral (sagittal), and the vein is a nearby collateral vessel with pressure equal to zero, if the venous end is protected by a normal pair of valves (so that there is no venous hypertension). This type of anastomosis means a very small wound site and ensures that a very small biopsy can be performed involving all nodal planes at the time of preparation for the anastomosis. This offers adequate information on the status or involvement of lymph nodes in these primary lymphedemas. If there is venous hypertension in the vessels near the proposed level of anastomosis, we first plan an appropriate shunt using Palma’s technique,82 or we create new circuits, as for the Cockett or May-Thurner syndrome,83 and then create a vein lymph node anastomosis as a second step. If high local venous pressure means the procedure is likely to turn out unsatisfactorily, we make a Palma shunt for the anastomosis between a lymph node and the contralateral internal saphenous vein.84 The advantage is that this requires only one suture. In an upper limb, we translocate an external jugular vein and any of its collateral vessels for the lymph node–vein anastomosis at the axilla,84 and eventually the internal jugular vein to lessen venous hypertension in the limb, performing an end-to-end anastomosis with the subclavian vein. All patients undergoing such a surgical procedure also receive manual lymphatic drainage as necessary, and during the daytime, customized soft compression nonelastic support and taping are applied. We use this alternative technique occasionally in cancer patients who have undergone radical surgery, and in some patients with Klippel-Trenaunay-Servelle syndrome with agenesia or extreme segmental hypoplasia of the subclavian vein.

Conclusion It seems that the best outcomes are gained in the treatment of pediatric lymphedema when we consider and use the clearly established guidelines of the international consensus documents. These can provide core recommendations that are then augmented by information from a comprehensive patient and family assessment.

K23346_Neligan_16_Lymph in Ped_r3_dc_0235-0246.indd 242

5/26/2015 9:11:07 AM

Chapter 16  Lymphedema in Pediatric Patients

243

In all cases of pediatric lymphedema it is important to realize that the parents are young when they are told about this significant and unexpected disorder in their child. Care of a child with primary lymphedema is an ongoing process involving significant parent cooperation and the stresses attendant on this care can have a deleterious effect on the couple’s relationship, and family stability can become compromised. Often treatment and management needs to extend to the whole family, requiring interaction with and assistance from a wide range of health care professionals. Lymphedema in pediatric patients requires a treatment plan that is quite different from a regimen designed for adults, in large measure because the reasons for the condition are generally different. The dominant concerns in pediatric patients—malformations and truncular venous hypertension and all considerations on the subject of growth and lymphedema—are typical of childhood, and these young patients are faced with living with the disorder for approximately eight decades. Given the need for different therapeutic approaches, it is necessary to identify and group the different disorders (diseases or syndromes). We have much to learn about the underlying reasons for and consequences of lymphatic malformation and thus how we should target treatment.

C linical P earls • To achieve the best outcomes, a comprehensive objective and subjective assessment of the patient is essential and must include a full family history as its basis. • Pediatric lymphedema requires more time from the physician, a dedicated health care space, specific psychophysical considerations, and a regard for the associated complex social aspects of the lymphedema on patient and family dynamics. • Hypoplasias and hyperplasias occur more frequently than indicated in the literature about primary lymphedemas, and health care providers should be knowledgeable and prepared to recognize and treat these.

R EFERENCES 1. Papendieck CM. Linfedema en pediatría. Clasificacion y etiopatogenia. Rev Hosp Niños BAires 45(201):14-22, 2003. 2. WHO Expert Committee on Lymphatic Filariasis. 1983 FIL7EC7WWP782.23 TDR. 3. Bancroft J. Cases of filarious disease. Trans Pathol Soc (London) 29:407-410, 1878. 4. Niño FL. Parasitologia. Puebla, Mexico: JM Cajica Publisher, 1958. 5. Chernin E, Manson P. The transmission of filariasis. Am J Trip Med Hyg 261:1065-1070, 1977. 6. Manson P. Elephantiasis arabum in the South Sea Islands. Br Med J 1(1744):1186-1187, 1894. 7. Romaña C, Wygodzinsky P. Acerca de la transmisíon de la Mansonella ozzardi (Filaria tucumana) Biglioeri y Araoz. An Inst Med Reg 3:29-34, 1950. 8. Papendieck CM. Lymphatic dysplasias in paediatrics. A new classification. Intern Angiol 18:6-9, 1999. 9. Földi M, Földi E, eds. Földi’s Textbook of Lymphology for Physicians and Lymphedema Therapists, ed 3. St Louis: Mosby Elsevier, 2012. 10. Papendieck CM. Lymphangioadenodysplasias in paediatrics. Lymphology 29:27-29, 1996. 11. Földi M, Földi E, Kubik S. Lehrbuch der Lymphologie, ed 6. Munich: Elsevier, 2005.

K23346_Neligan_16_Lymph in Ped_r3_dc_0235-0246.indd 243

5/26/2015 9:11:07 AM

244

Part III  Pathophysiology and Clinical Presentation

12. Papendieck CM. Lymphangioadenodysplasias. Presented at the Twenty-fourth Meeting of the European Group of Lymphology (GEL), Fifth Meeting of the Latin Mediterranean Chapter of the International Society of Lymphology, San Marco di Castellabate, Salerno, 1998. 13. Nikitenko L, Shimosawa T, Henderson S, et al. Adrenomedullin haploinsufficiency predisposes to secondary lymphedema. J Invest Dermatol 133:1768-1776, 2013. 14. Witte MH, Dellinger MT, Bernas M, Jones KA, Witte CH. Molecular lymphology and genetics of lymphedema: angiodysplasia syndromes. In Földi M, ed. Földi’s Textbook of Lymphology for Physicians and Lymphedema Therapists, ed 2. St Louis: Mosby Elsevier, 2006. 15. Moffatt C. International Consensus Best Practice for the Management of Lymphoedema. LF. MEP2006. 6-8. 16. Temple K. Distichiasis-lymphedema. Clin Dysmorphol 3:139-142, 1994. 17. Witte MH, Dellinger MT, Bernas MJ, et al. Molecular lymphology and genetics of lymphedema-angiodysplasia syndromes. In Földi M, Földi E, eds. Földi’s Textbook of Lymphology, ed 2. St Louis: Mosby Elsevier, 2006. 18. Brorson H, Ohlin K, Olsson G, et al. Adipose tissue dominates chronic arm lymphedema following breast cancer: an analysis using volume rendered CT images. Lymphat Res Biol 4:199-210, 2006. 19. Ryan TJ. Lymphatics and adipose tissue. Clin Dermatol 13:493-498, 1995. 20. Hennekam RCM. Lymphatic syndromic maldevelopment. Presented at the Fourth National Lymphedema Network (NLN) International Conference, Orlando, FL, 2000. 21. Nordkild P, Kromann-Andersen H, Struve-Christensen E. Yellow nail syndrome—the triad of yellow nails, lymphedema and pleural effusions. A review of the literature and a case report. Acta Med Scand 219:221-227, 1986. 22. Papendieck CM, Bergada C, Gruñeiro L. Noonan syndrome with generalized lymphangiectasia. Rev Hosp Niños BAires 62:20-22, 1974. 23. Calzada LR. Identificacion y Nanejo del Niño con Talla Baja: Sindrome de Ullrich Turner. Mexico: Intersistema, 2007. 24. Allanson JE. Noonan syndrome. J Med Genet 24:9-13, 1987. 25. Somer M. Diagnostic criteria and genetics of the PEHO syndrome. J Med Genet 30:932-936, 1993. 26. Bronspiegel N, Zelnick N, Rabinowitz H, et al. Aplasia cutis congenita and intestinal lymphangiectasia. An unusual association. Am J Dis Child 139:509-513, 1985. 27. Carchon H, Van Schaftingen E, Matthijs G, et al. Carbohydrate-deficient glycoprotein syndrome type IA (phosphomannomutase-deficiency). Biochim Biophys Acta 1455:155-165, 1999. 28. Dobyns WB, Patton MA, Stratton RF, et al. Cobblestone lissencephaly with normal eyes and muscle. Neuropediatrics 27:70-75, 1996. 29. Har-El G, Borderon ML, Weiss MH. Choanal atresia and lymphedema. Ann Otol Rhinol Laryngol 100:661-664, 1991. 30. Aagenaes O. Hereditary cholestasis with lymphoedema (Aagenaes syndrome, cholestasis-lymphoedema syndrome). New cases and follow-up from infancy to adult age. Scand J Gastroenterol 33:335-345, 1998. 31. Wässer ST, Beyreiss K, Himmel D, et al. Exsudative Enteropathie bei kongenitaler intestinaler lymphangiektasie mit multipler Skelettveränderung. Acta Paediatr Acad Sci Hung 15:17-25, 1974. 32. Urioste M, Rodríguez JI, Barcia JM, et al. Persistence of müllerian derivatives, lymphangiectasis, hepatic failure, postaxial polydactyly, renal and craniofacial anomalies. Am J Med Genet 47:494-503, 1993. 33. Avasthey P, Roy SB. Primary pulmonary hypertension, cerebrovascular malformation, and lymphoedema feet in a family. Br Heart J 30:769-775, 1968. 34. Figueroa AA, Pruzansky S, Rollnick BR. Meige disease (familial lymphedema praecox) and cleft palate: report of a family and review of the literature. Cleft Palate J 20:151-157, 1983. 35. Dahlberg PJ, Borer WZ, Newcomer KL, et al. Autosomal or X-linked recessive syndrome of congenital lymphedema, hypoparathyroidism, nephropathy, prolapsing mitral valve, and brachytelephalangy. Am J Med Genet 16:99-104, 1983.

K23346_Neligan_16_Lymph in Ped_r3_dc_0235-0246.indd 244

5/26/2015 9:11:07 AM

Chapter 16  Lymphedema in Pediatric Patients

245

36. Emberger JM, Navarro M, Dejean M, et al. [Deaf-mutism, lymphedema of the lower limbs and hematological abnormalities (acute leukemia, cytopenia) with autosomal dominant transmission] J Genet Hum 27:237-245, 1979. 37. Crowe CA, Dickerman LH. A genetic association between microcephaly and lymphedema. Am J Med Genet 24:131-135, 1986. 38. Limwongse C, Wyszynski RE, Dickerman LH, et al. Microcephaly-lymphedema-chorioretinal dysplasia: a unique genetic syndrome with variable expression and possible characteristic facial appearance. Am J Med Genet 86:215-218, 1999. 39. Opitz JM, Reynolds JF, FitzGerald JM. Mandibulofacial dysostosis or bilateral hemifacial microsomia with hearing loss, telecanthus, tetramelic postaxial hexadactyly, congenital hypotonia and lymphedema with joint hypermobility, and pigmentary dysplasia: a new syndrome? Am J Med Genet 33:433-435, 1989. 40. Alexander IE, Tauro GP, Bankier A. Fetal brain disruption sequence in sisters. Eur J Pediatr 154:654657, 1995. 41. Cormier-Daire V, Lyonnet S, Lehnert A, et al. Craniosynostosis and kidney malformation in a case of Hennekam syndrome. Am J Med Genet 57:66-68, 1995. 42. Mücke J, Hoepffner W, Scheerschmidt G, et al. Early onset lymphoedema, recessive form—a new form of genetic lymphoedema syndrome. Eur J Pediatr 145:195-198, 1986. 43. Dumić M, Vukelić D, Cviko A, et al. Nevo syndrome. Am J Med Genet 76:67-70, 1998. 44. Hennekam RC, Geerdink RA, Hamel BC, et al. Autosomal recessive intestinal lymphangiectasia and lymphedema, with facial anomalies and mental retardation. Am J Med Genet 34:593-600, 1989. 45. Klippel M, Trenaunay P. Du naevus variquex osteohypertrophique. Arch Gen Med 3:641-672, 1900. 46. Papendieck CM. Sindrome de Klippel Trenaunay Servelle en pediatria. Rev Arg Cirugia 64:42-44, 1993. 47. Papendieck CM, Barbosa ML, Pozo P, et al. Klippel-Trenaunay-Servelle syndrome in pediatrics. Lymphat Res Biol 1:81-85, 2003. 48. Weber FP. Haemangiectasic Hypertrophy of the Foot and Lower Extremity, Congenital or Acquired. London: Presse Med, 1918. 49. Wiedemann HR. The proteus syndrome. Eur J Pediatr 140:5, 1983. 50. Papendieck CM. El sindrome proteo en pediatria. Prensa Med Arg 85:348-351, 1998. 51. Gucev ZS, Tasic V, Jansevska A, et al. Congenital lipomatous overgrowth, vascular malformations, and epidermal nevi (CLOVE) syndrome: CNS malformations and seizures may be a component of this disorder. Am J Med Genet A 146A:2688-2690, 2008. 52. Gorham WL, Stout PH. Massive osteolysis (acute spontaneous absorption of bone, phantom bone disease, disappearing bone). J Bone Joint Surg Am 37:985-1004, 1955. 53. Mulliken JB, Young AE. Vascular Birthmarks: Hemangiomas and Malformations. Philadelphia: WB Saunders, 1988. 54. Papendieck CM. Sindrome de Gorham Stout Haferkamp con reflujo de quilo. Rev Arg Cirugia 79:710, 2000. 55. Aupali T, Ismid IS, Wibowo H, et al. Evaluation of the prevalence of lymphatic filariasis. Trans R Soc Trop Med Hyg 100:753-759, 2006. 56. Price EW. Podoconiosis: Non Filarial Elephantiasis. The Elephantiasis Story, 1-6. Oxford, UK: Oxford Medical Publications, 1990. 57. Dewa G, Tekola F, Newport MJ. Podoconiosis, non-infectious geochemical elephantiasis. Trans R Soc Trop Med Hyg 101:1175-1180, 2007. 58. Price EW. Podoconiosis: The Geographical Distribution of Endemic Podoconiosis, 15-19. Oxford, UK: Oxford Medical Publications, 1990. 59. Price EW. Podoconiosis: Pathogenesis and Pathology, 61-84. Oxford, UK: Oxford Medical Publications, 1990. 60. Kluken N. Documenta Angiologorum Vol XXIII. Curatorium Angiologiae Internationalis Ratschow Award 1993: Papendieck CM. Zur Bedeutung der Hypertension des Venen Systems im Kindesalter, 6-16, 1993.

K23346_Neligan_16_Lymph in Ped_r3_dc_0235-0246.indd 245

5/26/2015 9:11:07 AM

246

Part III  Pathophysiology and Clinical Presentation

61. Servelle M. Pathologie Vasculaire. Les Affections Veineuses. Malformations Congenitales des Veines des Members. Paris: Mason et Cie, 1978. 62. Papendieck CM. The big angiodysplastic syndromes in pediatrics with the participation of the lymphatic system. Progress in Lymphology XVI. Lymphology 31(Suppl):390-392, 1998. 63. Berry SA, Pearson C, Mize W, et al. Klippel-Trenaunay syndrome. Am J Med Genet 79:319-326, 1998. 64. Papendieck CM. Angiodysplasias in Pediatrics: Klippel-Trenaunay Syndrome. Buenos Aires, Argentina: Médica Panamericana, 1988. 65. Papendieck CM. Pediatric Angiology: Klippel-Trenaunay-Servelle Syndrome. Buenos Aires, Argentina: Médica Panamericana, 1992. 66. Wiedemann HR, Burgio GR, Aldenhoff P, et al. The proteus syndrome. Partial gigantism of the hands and/or feet, nevi, hemihypertrophy, subcutaneous tumors, macrocephaly or other skull anomalies and possible accelerated growth and visceral affections. Eur J Pediatr 140:5-12, 1983. 67. Papendieck CM. El syndrome proteo en pediatria. Prensa Med Arg 85:348-351, 1998. 68. Sapp J, Turner J, Van de Kamp JM, et al. Newly delineated syndrome of congenital lipomatous overgrowth, vascular malformations, and epidermal nevi (CLOVE syndrome) in seven patients. Am J Med Genet A 143A:2944-2958, 2007. 69. Földi M, Földi E. The hair tourniquet syndrome in the newborn. In Földi M, ed. Földi’s Textbook of Lymphology for Physicians and Lymphedema Therapists, ed 2. St Louis: Mosby Elsevier, 2006. 70. Benson CD, Mustard WT, Ravitch MM, et al. Cirugía Infantil [Pediatric Surgery] 2:1267-1268, 1967. 71. Karlberg J, Engstrom I, Karlbeg P, et al. Analysis of linear growth using a mathematical model. I. From birth to three years. Acta Paediatr Scand Suppl 76:478-488, 1987. 72. Karlberg J, Fryer J, Engstrom I, et al. Analysis of linear growth using a mathematical model. II. From 3 to 21 years of age. Acta Pediatr Scand Suppl 337:12-29, 1987. 73. Laguado Acevedo D, Villanueva N, Gonzalez D, et al. Integracion psicosocial de sindromes angiodisplasicos combinados en pediatria. Presented at the Thirty-ninth Congress of the European Society of Lymphology, Valencia, Spain, June 2013. 74. Bellini C, Ergaz M, Radicioni I, et al. Congenital fetal and neonatal visceral chylous effusions: neonatal chylothorax, and chylous ascites revisited. A multicenter retrospective study. Lymphology 45:91-102, 2012. 75. Boccardo F, Campisi C, Molinari L, et al. Diagnosis and treatment of chylous disorders. EJLRP 65:1519, 2012. 76. Servelle M, Nogues C. The Chyliferous Vessels. Paris: Expansion Scientifique Francaise, 1981. 77. Brorson H. Liposuction gives complete reduction of chronic large arm lymphedema after breast cancer. Acta Oncol 39:407-420, 2000. 78. Olszewski WL. Microsurgical lymphovenous anastomosis after 45 years’ follow up and indications. Presented at the Thirty-eighth Congress of the European Society of Lymphology, Berlin, June 2012. 79. Campisi C, Bellini C, Campisi C, et al. Microsurgery for lymphedema: clinical research and long-term results. Microsurgery 30:256-260, 2010. 80. Campisi C, Boccardo F. Microsurgical techniques for lymphedema treatment: derivative lymphaticvenous microsurgery. World J Surg 28:609-613, 2004. 81. Campisi C, Da Rin E, Bellini C, et al. Pediatric lymphedema and correlated syndromes: role of microsurgery. Microsurgery 28:138-142, 2008. 82. Palma EC, Esperon R. Vein transplant and grafts in the surgical treatment of the postphlebitic syndrome. J Cardiovasc Surg 1:94-107, 1960. 83. Cockett FB, Thoma M. The iliac vein compression syndrome. Br J Surg 52:816, 1965. 84. Papendieck CM. Venous bridges as an alternative option for lymphovenous shunts. Presented at the Thirty-seventh Congress of the European Society of Lymphology, Warsaw, Poland, June 2011.

K23346_Neligan_16_Lymph in Ped_r3_dc_0235-0246.indd 246

5/26/2015 9:11:07 AM

C hapter 17 Combined Lymphatic and Venous Failure: Phlebolymphedema Audra A. Duncan

Com ede K ey P oints • Lymphedema resulting from venous failure is frequently misdiagnosed. • A high frequency of lymphatic dysfunction is found in chronic venous disease. • If diagnosed, the edema may be reduced with venous outflow stenting and adjunctive measures, such as compression and risk factor modification. • Intravascular ultrasound is an effective way to image venous outflow obstruction not visualized on contrast venography.

Lymphedema caused by chronic venous disease is often characterized as secondary lymphedema and is often difficult to differentiate from primary lymphedema. In reality, the condition is best referred to as phlebolymphedema,1 because it represents a combined central (or local) venous insufficiency combined with lymphatic insufficiency. Other causes of secondary lymphedema include filariasis (which is the most common cause in tropical countries), surgical intervention associated with cancer treatment, radiation, or recurrent infection.2 If secondary causes are eliminated with a comprehensive evaluation of the patient with a swollen limb, the assumption is that the lymphedema is primary. However, what is often overlooked is the high frequency (20% to 30%) of lymphatic dysfunction found in patients with chronic venous disease. The major causes of lymphatic damage develop from an elevated capillary and venous pressure (resulting from either right-sided heart failure, immobility, or venous obstruction) and chronic fluid overload on the tissues.3-5 Lymphedema resulting from venous failure may improve with interventions designed to decrease venous hypertension, but only if the correct diagnosis is made. Therefore a deep understanding of the cause of limb swelling is needed to ensure that patients with lymphedema are diagnosed 247

K23346_Neligan_17_Phlebolymphedema_r3_0247-0254.indd 247

5/26/2015 9:23:14 AM

248

Part III  Pathophysiology and Clinical Presentation

with primary lymphedema only after a thorough evaluation. In this respect, it is essential that lymphologists and phlebologists work together in a team treatment capacity.6,7

Mechanism Primary or secondary lymphedema generally occurs as a result of the accumulation of interstitial (but not always) protein-rich fluid, whereas the edema noted in pure venous disease is generally low-protein. When venous insufficiency occurs, however, venous hypertension may overload the capacity of the lymphatics to drain from the limb, resulting in chronic lymphatic insufficiency. Thus a mixed lymphedema and venous edema results.

Diagnosis Limb swelling and/or cellulitis is the most common presentation of this combined failure, although lipodermatosclerosis, fibrosis and papillomatosis, and ulceration along the malleolus may also occur. However, in most patients in whom phlebolymphedema is unrecognized, the hallmark signs of venous hypertension, such as ankle hyperpigmentation and corona phlebectatica, although frequently seen, may not be obvious and sometimes lead to the misdiagnosis of primary lymphedema. In addition, clinical signs that are characteristic of lymphedema may be seen, such as pitting edema, squaring of the toes with swelling, and edema over the dorsum of the foot. In many patients, however, clinical findings are too nonspecific to make a definitive diagnosis without further imaging studies,8 although even the latter (MRI or CT) are often unable to provide a clear differentiation of the various forms of chronic edema.9

Noninvasive Studies Venous Plethysmography Venous plethysmography with air, strain-gauge, or photoplethysmography may be one of the early studies performed when evaluating the swollen limb. Typically findings are correlated with ultrasound for improved diagnostic sensitivity. Although studies have shown value in assessing venous functional changes and quantifying the results of surgical intervention,10,11 there is no clear role for venous plethysmography, in addition to duplex ultrasound.12

Imaging Duplex Imaging Although duplex imaging is often the primary imaging modality for both screening and the diagnosis of venous compression, it may not be sensitive enough to identify common iliac vein lesions. When performed in the upright and supine positions, duplex imaging may identify venous insufficiency or venous obstruction or reflux and quantify the degree of reflux. The criteria for valvular incompetence include a valve closure time greater than 1 second for femoral and popli-

K23346_Neligan_17_Phlebolymphedema_r3_0247-0254.indd 248

5/26/2015 9:23:14 AM

Chapter 17  Combined Lymphatic and Venous Failure: Phlebolymphedema

249

teal veins and 0.5 second for the great saphenous, small saphenous, tibial, deep femoral, and perforating veins.12 When ultrasound is used to identify outflow obstruction, however, the degree of iliac vein stenosis may be underestimated, and adjunctive imaging, such as CT, MR venography, or intravascular ultrasound (IVUS), may be necessary.8,13

Nucleotide Lymphangiography In both primary and secondary lymphedema, lymph node uptake may be delayed or absent, as demonstrated on nucleotide lymphangiography, even after prolonged observation periods.14,15 Lymphoscintigraphy may be more useful to assess improvement postoperatively, as will be discussed. When performed, an injection of technetium-99m sulfur colloid is given between the subcutaneous space of the first and second toes, and images are taken at scheduled intervals to assess movement of the colloid. The progress may be monitored for 2 hours or more, depending on the progress of the lymph drainage and the region of interest. The images are assessed for pooling of the tracer or reflux into the limb and the rate of movement up the limb (Fig. 17-1). Thus qualitative and quantitative information can be gained from the same imaging strategy. Typically, normal lymph drainage will allow movement of the colloid and visualization of the lymph nodes by 20 minutes.8 However, drainage is often affected by patient movement, facilitating lymph drainage (or not) during the observation period. Values still vary because of the lack of a standard protocol.9 Raju et al8 also recommend the use of triple-dose lymphangiography in which 1.8 mCi (compared with 0.6 mCi in standard lymphoscintigraphy) is injected to overcome the risk that a large fluid collection could dilute the isotope and interfere with lymphatic imaging. In their series, Raju et al8 report 71 limbs in all clinical, etiology, anatomy, pathophysiology (CEAP) classifications of chronic venous disease that had delayed or absent node visualization on standard lymphoscintigraphy. All 71 limbs underwent triple-dose lymphangiography. Of these 71 limbs, 10 were normal and 16 were abnormal in CEAP C3.8

Suspect mixed lymphedema/ venous failure Duplex scan

Equivocal

Venous reflux or obstruction

CTV or MRV

Lymphoscintigraphy Venogram 6 IVUS with iliac stent or ablation of reflux

FIG. 17-1  Algorithm for basic evaluation of a patient with suspected phleboedema. (CTV, CT venography; IVUS, intravascular ultrasound; MRV, MR venography.)

K23346_Neligan_17_Phlebolymphedema_r3_0247-0254.indd 249

5/26/2015 9:23:14 AM

250

Part III  Pathophysiology and Clinical Presentation

Computed Tomography or Magnetic Resonance Venography CT venography and MR venography are primarily used to identify venous outflow lesions, such as iliofemoral or iliocaval stenosis or occlusion. CT venography should be done in the venous delayed phase but may also be performed with injection into a vein of the affected limb. MR imaging with a gadolinium injection with T1-weighted spin echo, T2-weighted turbo spin echo, and fat-suppressed T2 images has the additional advantage of identifying fluid in the subcutaneous tissues. In addition, subfascial fluid collections, dermal edema, or fibrosis may be seen on MR venography. A honeycomb pattern in the subcutaneous tissue, particularly on coronal T1 images, along with fluid accumulation on T2 images, may be diagnostic of lymphedema.1

Venography and Use of Intravascular Ultrasound Contrast venography with the potential for intervention may be the most useful study to combine both the diagnosis of venous outflow obstruction and treatment concomitantly. In the series of Raju et al8 of 72 patients with CEAP C3 with abnormal lymphangiographic findings and limb swelling, venography had only a 61% sensitivity in the diagnosis of venous obstruction. In 57 venograms, 61% had an iliac vein lesion; collaterals were noted in 26% as opposed to 68 patients with IVUS, 63 of whom had an obstructive lesion.8,16 However, IVUS may miss lesions that are adjacent to the hypogastric vein orifice, or focal and discrete.

Treatment and Results Because lymphedema as a consequence of chronic venous failure is a mixed etiologic disease, treatment modalities must address the underlying cause and its downstream effects.7 Although the treatment of venous hypertension may subsequently improve lymphatic drainage,13 it is often necessary to initiate nonsurgical treatment of lymphedema concomitantly. Venography with venous stenting can be used to treat iliac and caval outflow obstruction. If reflux is the main etiologic factor, endovenous treatment of reflux of the great saphenous vein with ablation (radiofrequency ablation or laser therapy) or foam sclerotherapy can be considered. Every effort should be made to use endovascular or endovenous means to treat phleboedema to minimize lymphatic dysfunction. If open surgical techniques are required (for example, venous outflow procedures such as the Palma procedure), further lymphatic complications may occur because of the disruption of already failing lymphatics.1 In a series by Szuba et al,17 in 17 patients with lymphedema of the upper and lower extremities, 15 of 17 cases of edema improved after interventional treatment of venous outflow obstruction. Neglen et al18,19 reported stent placement was typically done in their patients for all venous stenoses of the iliac veins, with stents ranging from 8 to 14 mm. Self-expanding, conformable stents are most commonly chosen for treating venous outflow obstruction.17-19

K23346_Neligan_17_Phlebolymphedema_r3_0247-0254.indd 250

5/26/2015 9:23:14 AM

Chapter 17  Combined Lymphatic and Venous Failure: Phlebolymphedema

251

To assess postoperative outcomes, patients are assessed by changes in CEAP classification, pain, sleep parameters, and objective swelling. Quality of life data with validated forms, such as the Chronic Venous Disease Quality of Life Questionnaire (CIVIQ), may provide insight into the success of the intervention. To quantify outcomes more accurately, Raju et al13 used preoperative and postoperative lymphoscintigraphy. In their series of 26 patients with lower limb swelling, preoperative technetium-99m sulfur colloid lymphoscintigraphy was abnormal (absent in 8 patients and reduced in 18). All patients had venous outflow stenosis based on duplex ultrasound and/or IVUS. After venoplasty and vein stenting in all 26 patients, follow-up lymphoscintigraphic findings were normal in 10 limbs, improved in 9, and remained the same in 7. At clinical follow-up at a mean of 1 year, 16 of 26 limbs were clinically improved, and 6 were completely asymptomatic for swelling. Correspondingly, many patients had improvement in pain symptoms, including nine who were completely pain free.13

Compression The use of compression may be critical, because many patients will not improve, even with the treatment of venous outflow obstruction resulting from long-standing lymphatic disease. Compression with low-stretch elastic wrapping, such as those used in lymphatic nonsurgical treatment, provides resistance when the limb moves, thereby mimicking a pressure gradient on the limb. Although high-stretch bandages (40 to 50 mm Hg for lymphatic disease and 30 to 40 mm Hg for venous disease) have been recommended in the past, the use of multilayered bandages has proved superior over single-layer compression in recent studies.20 Regardless of the type of compression, in the presence of venous ulcers, healing is improved with the use of compression compared with no compression therapy.12,21

Other Factors in the Management of Mixed Lymphedema and Venous Disease As with all patients with lymphedema, risk factor modification to minimize symptoms is an excellent adjunct to compression and venous interventions. Techniques include: • Exercising to improve calf muscle pump function, thereby increasing lymphatic limb flow • Performing range-of-motion and stretching exercises • Elevating the limb above the level of the heart both during the day and at night in bed • Maintaining a healthy weight • Avoiding pressure to the affected limb • Maintaining excellent skin hygiene Patients often ask about the role of medications in the treatment of edema. Diuretics are often tried in the early development of lymphedema or venous edema, but they do not provide a permanent solution.

K23346_Neligan_17_Phlebolymphedema_r3_0247-0254.indd 251

5/26/2015 9:23:14 AM

252

Part III  Pathophysiology and Clinical Presentation

Conclusion Lymphedema caused by venous failure may occur in 20% to 30% of patients but is often misdiagnosed. When it is diagnosed, relief of edema may occur with venous outflow stenting or the management of venous reflux, along with adjunctive measures to control limb swelling. If venous failure is suspected but a cause is not readily identified, IVUS may help in imaging venous outflow obstruction that is not visualized on venography.

C linical P earls • The venous contribution to limb edema should be considered early in diagnosis and management. • IVUS can be used to uncover venous stenosis that is not imaged on contrast venography. • If venous outflow obstruction is diagnosed, both the venous disease and lymphedema should be treated concomitantly. • Adjuvant risk factor modification such as exercise and limb elevation are important keys to treatment.

R EFERENCES 1. Bunke N, Brown K, Bergan J. Phlebolymphedema: usually unrecognized, often poorly treated. Perspect Vasc Surg Endovasc Ther 21:65-68, 2009. 2. Földi M. Classification of lymphedema and elephantiasis. Presented at the Twelfth International WHO/ TDR/FIL Conference on Lymphatic Pathology and Immunopathology in Filariasis, Thanjavur, India. Lymphology 18:148-168, 1985. 3. Bull RH, Gane JN, Evans JE, et al. Abnormal lymph drainage in patients with chronic venous leg ulcers. J Am Acad Dermatol 28:585-590, 1993. 4. Keely V. The use of lymphoscintigraphy in the management of chronic lymphoedema. J Lymphoedema 1:42-57, 2008. 5. Gloviczki P, Calcagno D, Schirger A, et al. Noninvasive evaluation of the swollen extremity: experiences with 190 lymphoscintigraphic examinations. J Vasc Surg 9:683-689; discussion 690, 1989. 6. Partsch H, Lee BB. Phlebology and lymphology—a family affair. Phlebology 29:645-647, 2014. 7. Piller N. Phlebolymphoedema/chronic venous lymphatic insufficiency: an introduction to strategies for detection, differentiation and treatment. Phlebology 24:51-55, 2009. 8. Raju S, Furth JB IV, Neglen P. Diagnosis and treatment of venous lymphedema. J Vasc Surg 55:141-149, 2012. 9. Weissleder H. Diagnosis of lymphostatic oedema of the extremities. Fortschr Med 115:32-36, 1997. 10. Green S, Thorp R, Reeder EJ. Venous occlusion plethysmography versus Doppler ultrasound in the assessment of leg blood flow during calf exercise. Eur J Appl Physiol 111:1889-1900, 2011.

K23346_Neligan_17_Phlebolymphedema_r3_0247-0254.indd 252

5/26/2015 9:23:14 AM

Chapter 17  Combined Lymphatic and Venous Failure: Phlebolymphedema

253

11. Weingarten MS, Czeredarczuk M, Scovell S, et al. A correlation of air plethysmography and color flowassisted duplex scanning in the quantification of chronic venous insufficiency. J Vasc Surg 24:750-754, 1996. 12. O’Donnell TF Jr, Passman MA, Marston WA, et al; Society for Vascular Surgery; American Venous Forum. Management of venous leg ulcers: clinical practice guidelines of the Society for Vascular Surgery® and the American Venous Forum. J Vasc Surg 60(2 Suppl):3S-59S, 2014. 13. Raju S, Owen S Jr, Neglen P. Reversal of abnormal lymphoscintigraphy after placement of venous stents for correction of associated venous obstruction. J Vasc Surg 34:779-784, 2001. 14. Gloviczki P, Calcagno D, Schirger A, et al. Noninvasive evaluation of the swollen extremity: experiences with 190 lymphoscintigraphic examinations. J Vasc Surg 9:683-689; discussion 690, 1989. 15. Browse N, Burnand KG, Mortimer P. Diseases of the Lymphatics. London: Arnold, 2003. 16. Neglen P, Thrasher TL, Raju S. Venous outflow obstruction: an underestimated contributor to chronic venous disease. J Vasc Surg 38:879-885, 2003. 17. Szuba A, Razavi M, Rockson SG. Diagnosis and treatment of concomitant venous obstruction in patients with secondary lymphedema. J Vasc Interv Radiol 13:799-803, 2002. 18. Neglen P, Raju S. Balloon dilation and placement of venous stent of chronic iliac vein obstruction: technical aspects and early clinical outcome. J Endovasc Ther 7:79-91, 2000. 19. Neglen P, Berry MA, Raju S. Endovascular surgery in the treatment of chronic primary and postthrombotic iliac vein obstruction. Eur J Vasc Endovasc Surg 20:560-571, 2000. 20. O’Meara S, Tierney J, Cullum N, et al. Four layer bandage compared with short stretch bandage for venous leg ulcers: systematic review and meta-analysis of randomised controlled trials with data from individual patients. BMJ 338:b1344, 2009. 21. O’Meara S, Cullum N, Nelson EA, et al. Compression for venous leg ulcers. Cochrane Database Syst Rev 11:CD000265, 2012.

K23346_Neligan_17_Phlebolymphedema_r3_0247-0254.indd 253

5/26/2015 9:23:14 AM

K23346_Neligan_17_Phlebolymphedema_r3_0247-0254.indd 254

5/26/2015 9:23:14 AM

C hapter 18 Lymphedema Risk Factors in Breast Cancer Swetha Kambhampati, Stanley Rockson

K ey P oints

P Lym

• Breast cancer treatment, one of the most common causes of lymphedema, affects more than one in five breast cancer survivors. • The presence of lymphedema has a substantial impact on a person’s physical, psychological, and social wellbeing. • Axillary surgery is one of the major risk factors for lymphedema. • The risk of lymphedema is not completely eliminated with sentinel node biopsy. • Postoperative radiation increases the risk of lymphedema by as much as 10-fold compared with surgery alone. • Exercise is beneficial. • Air travel, an elevated BMI, advanced age, and venous obstruction may contribute to the appearance or worsening of lymphedema.

Breast cancer treatment is one of the most common causes of lymphedema; more than one in five women who survive breast cancer will subsequently develop this chronic disease. Lymphedema after breast cancer treatment is characterized by regional swelling in the arm(s) ipsilateral to axillary staging and therapy; the acquired lymphatic vascular insufficiency leads to an accumulation of protein-rich interstitial fluid in the tissues distal to the obstructive lesion.1,2 This high incidence of lymphedema has been linked to treatment variables, along with a variety of other stressors that are still under active investigation. The cumulative incidence of arm lymphedema increases over time and is most pronounced during the initial 2 years after breast cancer intervention.3,4 Lymphedema has an extensive negative impact on the affected individual, including the physical symptoms of discomfort, pain, and limb tightness, accompanied by the loss of function in the limb. This functional deficit can compromise the individual’s work and social productivity and impact the ability to undertake self-care.5 The disease produces various pathologic sequelae, including an increased risk of infection6-9 and secondary malignancy.10-13 The negative impact on social 255

K23346_Neligan_18_Risk in Cancer_r2_0255-0262.indd 255

5/26/2015 9:12:18 AM

256

Part III  Pathophysiology and Clinical Presentation

relationships, emotional self-confidence, and psychological well-being cannot be overstated.14,15 The high incidence of lymphedema in breast cancer survivors, and its substantial impact on quality of life, suggest that the study of lymphedema and its associated risk factors is of high public health importance.

Treatment-Related Risk Factors for the Development of Lymphedema Patients who are in the early stages of breast cancer typically undergo primary surgery on the breast (lumpectomy or mastectomy) and regional nodes, with or without radiotherapy. After this local therapy, the patient may undergo systemic adjuvant therapy, depending on the tumor characteristics. Patients may choose to undergo either breast-conserving therapy (BCT), which consists of a lumpectomy plus radiotherapy, or a total mastectomy. A meta-analysis of 72 studies3 strongly supported an association between axillary lymph node dissection, the extent of lymph nodes dissected, and mastectomy and the development of lymphedema. There is moderate evidence to support an association between the risk of lymphedema after treatment and a higher number of lymph nodes with metastasis and the use of radiotherapy and chemotherapy. There is less robust support for a link between lymphedema development and the use of axillary radiotherapy.3 The diagnostic and treatment approaches to the management of breast cancer can have a significant impact on the risk of lymphedema after treatment, and an understanding of this can help physicians counsel patients appropriately.

Axillary Lymph Node Dissection Axillary surgery is one of the major risk factors for lymphedema. A desire to minimize the adverse outcomes associated with axillary lymph node dissection (ALND) led to the development of the sentinel lymph node biopsy (SLNB) technique. SLNB as a standard means of axillary nodal assessment significantly reduced the number of women undergoing axillary surgery and thus the prevalence of breast cancer treatment–induced lymphedema.16 A recent analysis suggests that the risk of lymphedema within 1 year of treatment is 2% after SLNB alone compared with 13% after SLNB combined with ALND.17 Another meta-analysis of lymphedema after breast cancer showed that the incidence of lymphedema in women who underwent ALND was almost four times higher than in those women who had SLNB.3 Because of the decreased adverse outcomes with SLNB, the American College of Surgeons Oncology Group initiated the Z0011 trial in 1999 to assess whether ALND improved outcomes in patients with sentinel node metastases who were undergoing BCT. The study demonstrated that SLNB alone did not result in inferior survival, a finding that has the potential to significantly change clinical practice.17 Based on this study, it is assumed that only patients with clinically suspicious axillary lymph nodes and a positive biopsy finding should undergo ALND at the time of BCT surgery. Other indications for ALND include patients with positive sentinel nodes undergoing mastectomy without postsurgical radiotherapy or patients with more than three positive sentinel nodes who are undergoing BCT. Patients with a negative biopsy result or clinically

K23346_Neligan_18_Risk in Cancer_r2_0255-0262.indd 256

5/26/2015 9:12:18 AM

Chapter 18  Lymphedema Risk Factors in Breast Cancer

257

negative axillary examination with no suspicious palpable axillary nodes should only undergo SLNB at the time of BCT surgery. This will help the surgeon make decisions regarding adjuvant systemic therapy and radiotherapy while minimizing the potential for complications and adverse outcomes, such as lymphedema. However, even with the SLNB technique, the risk of lymphedema is not completely eliminated.18,19 It is not only axillary surgery but even more limited breast-conserving interventions (with or without local radiotherapy) that can increase the risk of developing lymphedema.20,21 A study comparing patients with ALND and radical mastectomy with patients with ALND and BCT followed by local radiotherapy revealed that the incidence of lymphedema with mastectomy was more than twice that of the other group.22

Radiotherapy Although surgical interventions significantly increase the risk that lymphedema will develop, the use of adjunctive radiotherapy in breast cancer treatment further increases this risk.23 Postoperative radiotherapy increases the risk by as much as 10 times compared with surgery alone. A possible explanation for this effect may reside in the impact of radiation on the residual lymphatic structures within the radiation port. Radiation may also promote tissue fibrosis, thereby suppressing lymphatic regeneration.24 The inclusion of the supraclavicular or posterior axillary regions within the target radiation field increases the risk of lymphedema twofold or threefold, respectively. To minimize the risk of lymphedema induced by radiotherapy used as an adjunct to surgical breast cancer treatment, radiation oncologists have adopted newer radiotherapy protocols and three-dimensional methods that minimize normal tissue exposure and conventional fractionation to the involved nodal regions.23,25,26 This allows collateral lymphatic drainage and reduces the risk of lymphedema.23

Chemotherapy Traditionally it was assumed that chemotherapy did not increase the risk of lymphedema.27 However, more recent studies have shown an increased risk of lymphedema after chemotherapy for breast cancer. This association between lymphedema and chemotherapy may exist because systemic chemotherapy is often used in more advanced stages of the disease requiring more extensive surgical treatment, which has a clear link to a higher incidence of lymphedema. The relationship between chemotherapy and lymphedema may also exist because more pharmacotherapy and chemotherapeutic agents are used today than in the past, resulting in an increase in adverse interactions between these agents and the lymphatic system, thereby leading to lymphedema.3,28,29

Nontreatment-Related Risk Factors for the Development of Lymphedema For many years there has been controversy regarding the impact of other risk factors on the incidence of lymphedema in breast cancer survivors, which resulted in much fear and frustration in the patient population.30 Physicians long discouraged physical exercise of the affected limb

K23346_Neligan_18_Risk in Cancer_r2_0255-0262.indd 257

5/26/2015 9:12:18 AM

258

Part III  Pathophysiology and Clinical Presentation

with lymphedema in breast cancer patients because they believed that exercise would precipitate lymphedema in these patients or exacerbate the already extant limb swelling.31 However, more recent studies and trials have challenged this belief and demonstrated that exercise does not worsen lymphedema.32-36 On the contrary, the available evidence supports the concept that exercise can help control and improve lymphedema in these patients.3,5,37-39 Airplane travel has also traditionally been thought to worsen lymphedema. A single-subject case study showed that interarm bioimpedance ratios (a measure of interstitial fluid volume disparities) worsened after flying.38 A larger study used questionnaires to assess the role of aircraft travel in precipitating lymphedema or worsening the existing condition in patients who were treated for breast cancer; the observed relationship led to the hypothesis that the lowered aircraft cabin pressure can compromise the lymphatic function in the limb, either through obstruction of veins and lymphatics or diminished lymphatic pumping, thus leading to an accumulation of fluid in the limb.40,41 However, given this mechanism, it can be conjectured that patients can reduce the flight-associated risk with the use of additional compression (for example, inflated splints or pressure bandages) during the flight.40 Some studies contradict this general notion that air travel may trigger lymphedema. For example, one study observed athletic breast cancer survivors who traveled from Canada to Australia for a dragon boat competition without a demonstrable adverse relationship between air travel and arm lymphedema.42 Air travel alone may not be enough to precipitate or worsen lymphedema in breast cancer patients after treatment but may be influenced by other factors such as exercise, which may confer a protective effect on the patients’ inherent predisposition to develop lymphedema. Certain stressors, such as air travel, may impose a significant load on an already stressed lymphatic system and thus may precipitate the lymphedema or induce an acute flare in the swelling.30 Obesity is also considered a risk factor for lymphedema. Many studies suggest that a BMI greater than 25 or a sedentary lifestyle can increase the likelihood of developing lymphedema in breast cancer survivors.43-47 Hypertension may also be an important comorbidity that predisposes patients with a history of breast cancer treatment to develop lymphedema. An infection of the affected arm can also increase the risk of lymphedema in this patient population.43,48,49 More recent observations indicate that venous disruption in patients after mastectomy and radiation can also play a role in the pathogenesis of lymphedema. In a study of 81 patients, more than half had venous outflow obstruction on Doppler imaging of the venous flow of the upper extremity.50 Our own case series suggests that venous outflow obstruction not only precipitates lymphedema, but also renders the swelling refractory to decongestive therapy.51 This conclusion is further substantiated by the therapeutic response seen in residual arm lymphedema after the venous obstruction is treated with percutaneous venoplasty, with or without stenting.51 Based on these findings, physicians have more opportunities to use nonsurgical techniques to help mitigate the swelling and enhance the responsiveness to conservative treatment of the lymphedema in patients with simultaneous venous flow abnormalities.

K23346_Neligan_18_Risk in Cancer_r2_0255-0262.indd 258

5/26/2015 9:12:18 AM

Chapter 18  Lymphedema Risk Factors in Breast Cancer

259

Inconclusive evidence exists to support other risk factors for lymphedema, such as demographic characteristics, dominant versus nondominant limb involvement, postoperative infection, and certain lifestyle-associated factors (for example, self-care, use of the affected arm, and education about lymphedema).3 Also, a growing body of evidence suggests that inflammation plays a key role in the pathogenesis of lymphedema.52-54 Although our understanding of lymphedema and the risk factors for precipitating or worsening this condition in breast cancer patients has increased significantly, there is still much that must be learned about the pathogenesis of this disease. Physicians who are knowledgeable about the many risk factors will be able to better educate their patients with a history of breast cancer on ways to significantly reduce their risk of developing posttreatment lymphedema. As a result, physicians will also dispel the myths and fears of these patients.

Risk Factors for Lymphedema Progression The onset of lymphedema is insidious. It presents with aching pain and a sense of heaviness, discomfort, and fullness in the affected limb before the development of any visually perceptible changes.55 The limb subsequently starts to swell. At this early stage, the edema is characterized as soft and pitting. The pitting nature of the edema at this stage reflects the retention of excess interstitial fluid in the tissues.2,56,57 As lymphedema progresses, this pitting quality tends to disappear, reflecting the development of fibrosis and adipose tissue deposition. Over time the skin becomes not only resistant to deformity by palpation, but also dry and firm with dermal thickening, cutaneous fibrosis, and hyperkeratotic skin.30,58 The subcutaneous and subfascial adipose tissue deposition increases as the lymphedema progresses to such a degree that by the advanced, nonpitting stage, 73% of the excess volume in the affected limb is composed of adipose tissue.30,59,60 In these later stages of lymphedema, patients develop restricted range of motion in the affected limb. Studies have shown that, in individuals predestined to develop lymphedema, the problem is very likely to develop within the first 2 years after treatment. Often symptoms are mild, with little perceptible swelling and less than a 2 cm interlimb circumferential difference.30 More than half of these women do not progress beyond this stage in the subsequent several years, but the remaining cohort of these affected individuals manifest continuously progressing and worsening lymphedema beyond the initial mild stage.55,61 One factor that contributes to the likelihood of lymphedema progression is the temporal duration of the edematous state.62 Although patient age does not increase the risk of developing lymphedema, it has a negative impact on the likelihood of lymphedema progression. Older patients are more likely to progress beyond the mild, early manifestations of lymphedema. Thus the trend toward an increasingly aging population can be predicted to have an adverse effect on the observed natural history of lymphedema. Cardiovascular conditions other than hypertension also predispose to the development of subcutaneous fibrosis seen in the later stages of lymphedema.

K23346_Neligan_18_Risk in Cancer_r2_0255-0262.indd 259

5/26/2015 9:12:18 AM

260

Part III  Pathophysiology and Clinical Presentation

C linical P earls • The diagnostic and treatment approaches to the management of breast cancer can have a significant impact on the risk of lymphedema after treatment, and this understanding can help physicians counsel patients appropriately. • Exercise can help prevent, control, and treat lymphedema. • Inflammation plays an important role in disease pathogenesis. • Advancing age has a negative impact on the severity of lymphedema after it is present.

R EFERENCES 1. Rockson SG. Diagnosis and management of lymphatic vascular disease. J Am Coll Cardiol 52:799-806, 2008. 2. Cheville AL, McGarvey CL, Petrek JA, et al. The grading of lymphedema in oncology clinical trials. Semin Radiat Oncol 13:214-225, 2003. 3. DiSipio T, Rye S, Newman B, et al. Incidence of unilateral arm lymphoedema after breast cancer: a systematic review and meta-analysis. Lancet Oncol 14:500-515, 2013. 4. Schünemann H, Willich N. [Secondary lymphedema of the arm following primary therapy of breast carcinoma] Zentralbl Chir 117:220-225, 1992. 5. Hayes SC, Johansson K, Stout NL, et al. Upper-body morbidity after breast cancer: incidence and evidence for evaluation, prevention, and management within a prospective surveillance model of care. Cancer 118:2237-2249, 2012. 6. Vaillant L, Gironet N. [Infectious complications of lymphedema] Rev Med Interne 23 Suppl 3:403s407s, 2002. 7. Dupuy A, Benchikhi H, Roujeau JC, et al. Risk factors for erysipelas of the leg (cellulitis): case-control study. Br Med J 318:1591-1594, 1999. 8. Herpertz U. [Erysipelas and lymphedema] Fortschr Med 116:36-40, 1998. 9. Masmoudi A, Maaloul I, Turki H, et al. Erysipelas after breast cancer treatment (26 cases). Dermatol Online J 11:12, 2005. 10. Woodward AH, Ivins JC, Soule EH. Lymphangiosarcoma arising in chronic lymphedematous extremities. Cancer 30:562-572, 1972. 11. Stewart FW, Treves N. Classics in oncology: lymphangiosarcoma in postmastectomy lymphedema: a report of six cases in elephantiasis chirurgica. CA Cancer J Clin 31:284-299, 1981. 12. Tomita K, Yokogawa A, Oda Y, et al. Lymphangiosarcoma in postmastectomy lymphedema (StewartTreves syndrome): ultrastructural and immunohistologic characteristics. J Surg Oncol 38:275-282, 1988. 13. Cozen W, Bernstein L, Wang F, et al. The risk of angiosarcoma following primary breast cancer. Br J Cancer 81:532-536, 1999. 14. Tobin MB, Lacey HJ, Meyer L, et al. The psychological morbidity of breast cancer-related arm swelling. Psychological morbidity of lymphoedema. Cancer 72:3248-3252, 1993. 15. McWayne J, Heiney SP. Psychologic and social sequelae of secondary lymphedema: a review. Cancer 104:457-466, 2005. 16. D’Angelo-Donovan DD, Dickson-Witmer D, Petrelli NJ. Sentinel lymph node biopsy in breast cancer: a history and current clinical recommendations. Surg Oncol 21:196-200, 2012.

K23346_Neligan_18_Risk in Cancer_r2_0255-0262.indd 260

5/26/2015 9:12:18 AM

Chapter 18  Lymphedema Risk Factors in Breast Cancer

261

17. Giuliano AE, Hunt KK, Ballman KV, et al. Axillary dissection vs no axillary dissection in women with invasive breast cancer and sentinel node metastasis: a randomized clinical trial. JAMA 305:569-575, 2011. 18. Mansel RE, Fallowfield L, Kissin M, et al. Randomized multicenter trial of sentinel node biopsy versus standard axillary treatment in operable breast cancer: the ALMANAC Trial. J Natl Cancer Inst 98:599609, 2006. 19. Wilke LG, McCall LM, Posther KE, et al. Surgical complications associated with sentinel lymph node biopsy: results from a prospective international cooperative group trial. Ann Surg Oncol 13:491-500, 2006. 20. Stanisławek A, Kurylcio L, Janikiewicz A. Arm lymphoedema after surgical treatment for the cancer of the breast. Ann Univ Mariae Curie Sklodowska Med 55:155-160, 2000. 21. Kosir MA, Rymal C, Koppolu P, et al. Surgical outcomes after breast cancer surgery: measuring acute lymphedema. J Surg Res 95:147-151, 2001. 22. Nesvold IL, Dahl AA, Løkkevik E, et al. Arm and shoulder morbidity in breast cancer patients after breast-conserving therapy versus mastectomy. Acta Oncol 47:835-842, 2008. 23. Horst KC. Radiation complications. In Lee B, Bergan J, Rockson SG, eds. Lymphedema: A Concise Compendium of Theory and Practice. London: Springer, 2011. 24. Avraham T, Yan A, Zampell JC, et al. Radiation therapy causes loss of dermal lymphatic vessels and interferes with lymphatic function by TGF-beta1-mediated tissue fibrosis. Am J Physiol Cell Physiol 299:C589-C605, 2010. 25. Badiyan SN, Shah C, Arthur D, et al. Hypofractionated regional nodal irradiation for breast cancer: examining the data and potential for future studies. Radiother Oncol 110:39-44, 2014. 26. Graham P, Jagavkar R, Browne L, et al. Supraclavicular radiotherapy must be limited laterally by the coracoid to avoid significant adjuvant breast nodal radiotherapy lymphoedema risk. Australas Radiol 50:578-582, 2006. 27. Tsai RJ, Dennis LK, Lynch CF, et al. The risk of developing arm lymphedema among breast cancer survivors: a meta-analysis of treatment factors. Ann Surg Oncol 16:1959-1972, 2009. 28. Keser I, Basar S, Duzgun I, et al. Malpractice leading to secondary lymphedema after radical mastectomy: case report. Breast Care (Basel) 8:371-373, 2013. 29. Kilbreath SL, Lee MJ, Refshauge KM, et al. Transient swelling versus lymphoedema in the first year following surgery for breast cancer. Support Care Cancer 21:2207-2215, 2013. 30. Kilbreath SL. Breast cancer. In Lee B, Bergan J, Rockson SG, eds. Lymphedema: A Concise Compendium of Theory and Practice. London: Springer, 2011. 31. Lee TS, Kilbreath SL, Sullivan G, et al. Factors that affect intention to avoid strenuous arm activity after breast cancer surgery. Oncol Nurs Forum 36:454-462, 2009. 32. Harris SR, Niesen-Vertommen SL. Challenging the myth of exercise-induced lymphedema following breast cancer: a series of case reports. J Surg Oncol 74:95-98; discussion 98-99, 2000. 33. McKenzie DC, Kalda AL. Effect of upper extremity exercise on secondary lymphedema in breast cancer patients: a pilot study. J Clin Oncol 21:463-466, 2003. 34. Ahmed RL, Thomas W, Yee D, et al. Randomized controlled trial of weight training and lymphedema in breast cancer survivors. J Clin Oncol 24:2765-2772, 2006. 35. Turner J, Hayes S, Reul-Hirche H. Improving the physical status and quality of life of women treated for breast cancer: a pilot study of a structured exercise intervention. J Surg Oncol 86:141-146, 2004. 36. Bloomquist K, Karlsmark T, Christensen KB, et al. Heavy resistance training and lymphedema: prevalence of breast cancer-related lymphedema in participants of an exercise intervention utilizing heavy load resistance training. Acta Oncol 53:216-225, 2014. 37. Baumann FT, Bloch W, Weissen A, et al. Physical activity in breast cancer patients during medical treatment and in the aftercare—a review. Breast Care (Basel) 8:330-334, 2013. 38. Schmitz KH, Ahmed RL, Troxel A, et al. Weight lifting in women with breast-cancer-related lymphedema. N Engl J Med 361:664-673, 2009.

K23346_Neligan_18_Risk in Cancer_r2_0255-0262.indd 261

5/26/2015 9:12:18 AM

262

Part III  Pathophysiology and Clinical Presentation

39. Schmitz KH, Ahmed RL, Troxel AB, et al. Weight lifting for women at risk for breast cancer-related lymphedema: a randomized trial. JAMA 304:2699-2705, 2010. 40. Casley-Smith JR, Casley-Smith JR. Lymphedema initiated by aircraft flights. Aviat Space Environ Med 67:52-56, 1996. 41. Ward LC, Battersby KJ, Kilbreath SL. Airplane travel and lymphedema: a case study. Lymphology 42:139-145, 2009. 42. Kilbreath SL, Ward LC, Lane K, et al. Effect of air travel on lymphedema risk in women with history of breast cancer. Breast Cancer Res Treat 120:649-654, 2010. 43. Ugur S, Arici C, Yaprak M, et al. Risk factors of breast cancer-related lymphedema. Lymphat Res Biol 11:72-75, 2013. 44. Ahmed RL, Schmitz KH, Prizment AE, et al. Risk factors for lymphedema in breast cancer survivors, the Iowa Women’s Health Study. Breast Cancer Res Treat 130:981-991, 2011. 45. Kwan ML, Darbinian J, Schmitz KH, et al. Risk factors for lymphedema in a prospective breast cancer survivorship study: the Pathways Study. Arch Surg 145:1055-1063, 2010. 46. Helyer LK, Varnic M, Le LW, et al. Obesity is a risk factor for developing postoperative lymphedema in breast cancer patients. Breast J 16:48-54, 2010. 47. Hayes SC, Janda M, Cornish B, et al. Lymphedema after breast cancer: incidence, risk factors, and effect on upper body function. J Clin Oncol 26:3536-3542, 2008. 48. Shahpar H, Atieh A, Maryam A, et al. Risk factors of lymph edema in breast cancer patients. Int J Breast Cancer 2013:641818, 2013. 49. Ben Salah H, Bahri M, Jbali B, et al. [Upper limb lymphedema after breast cancer treatment] Cancer Radiother 16:123-127, 2012. 50. Svensson WE, Mortimer PS, Tohno E, et al. Colour Doppler demonstrates venous flow abnormalities in breast cancer patients with chronic arm swelling. Eur J Cancer 30A:657-660, 1994. 51. Szuba A, Razavi M, Rockson SG. Diagnosis and treatment of concomitant venous obstruction in patients with secondary lymphedema. J Vasc Interv Radiol 13:799-803, 2002. 52. Avraham T, Zampell JC, Yan A, et al. Th2 differentiation is necessary for soft tissue fibrosis and lymphatic dysfunction resulting from lymphedema. FASEB J 27:1114-1126, 2013. 53. Lin S, Kim J, Lee MJ, et al. Prospective transcriptomic pathway analysis of human lymphatic vascular insufficiency: identification and validation of a circulating biomarker panel. PloS One 7:e52021, 2012. 54. Leung G, Baggott C, West C, et al. Cytokine candidate genes predict the development of secondary lymphedema following breast cancer surgery. Lymphat Res Biol 12:10-22, 2014. 55. Norman SA, Localio AR, Potashnik SL, et al. Lymphedema in breast cancer survivors: incidence, degree, time course, treatment, and symptoms. J Clin Oncol 27:390-397, 2009. 56. Szuba A, Shin WS, Strauss HW, et al. The third circulation: radionuclide lymphoscintigraphy in the evaluation of lymphedema. J Nucl Med 44:43-57, 2003. 57. Stanton AW, Mellor RH, Cook GJ, et al. Impairment of lymph drainage in subfascial compartment of forearm in breast cancer-related lymphedema. Lymphat Res Biol 1:121-132, 2003. 58. Rockson SG. Lymphedema. Am J Med 110:288-295, 2001. 59. Brorson H, Ohlin K, Olsson G, et al. Breast cancer-related chronic arm lymphedema is associated with excess adipose and muscle tissue. Lymphat Res Biol 7:3-10, 2009. 60. Brorson H, Ohlin K, Olsson G, et al. Adipose tissue dominates chronic arm lymphedema following breast cancer: an analysis using volume rendered CT images. Lymphat Res Biol 4:199-210, 2006. 61. Bar Ad V, Cheville A, Solin LJ, et al. Time course of mild arm lymphedema after breast conservation treatment for early-stage breast cancer. Int J Radiat Oncol Biol Phys 76:85-90, 2010. 62. Casley-Smith JR. Alterations of untreated lymphedema and its grades over time. Lymphology 28:174185, 1995.

K23346_Neligan_18_Risk in Cancer_r2_0255-0262.indd 262

5/26/2015 9:12:18 AM

C hapter 19 Sentinel Lymph Node Biopsy Outcomes Janice N. Cormier, Kate D. Cromwell, Jane M. Armer, Merrick I. Ross

K ey P oints • Sentinel lymph node biopsy (SLNB) is an oncologically safe procedure to identify the primary lymph node to which a solid tumor, such as a melanoma, breast cancer, or gynecologic cancer, is likely to drain. • Compared with nodal observation alone, SLNB increases overall and disease-free survival in patients with intermediate-thickness tumors. • The incidence of lymphedema has been reduced but not eliminated by the use of the SLNB technique. • The incidence of lymphedema after SLNB ranges from 4% to 9% in the three tumor types (melanoma, breast cancer, and gynecologic cancer) discussed in this chapter. • Patient-reported quality of life is affected less by SLNB than by total lymphadenectomy.

The use of the sentinel lymph node biopsy (SLNB) has changed the landscape for the surgical treatment of many types of solid tumors, including melanomas and breast cancer. This minimally invasive procedure identifies and resects sentinel lymph nodes (SLNs) for the pathologic staging of lymph node basins in patients at risk for nodal metastases and can be used to avoid the morbidity associated with a complete lymphadenectomy in patients with pathologically confirmed negative SLNs.

P

This chapter outlines the technique for performing a SLNB and highlights recent literature findings related to the efficacy of the procedure in different types of cancers. Finally, there is a discussion of the impact of SLNB on lymphedema and quality of life (QOL) outcomes related to the use of this technique.

Sen

Gould et al1 first described the concept of SLNB in 1960 after an incidental finding of a lymph node at the junction of the anterior and posterior facial veins during a total parotidectomy. After node removal and identification of metastatic tumor cells, a complete cervical lymph node dissection was performed. In 1977 Cabanas2 published an article on SLNB at the time of surgical resection for penile cancer in which lymphatic imaging was used to identify the SLN. The SLN was then 263

K23349_Neligan_19_Sentinal Lymph_r5_dc_0263-0274.indd 263

5/27/2015 9:31:47 AM

264

Part III  Pathophysiology and Clinical Presentation

A

B

C

Regional lymph nodes

Sentinel lymph nodes Tumor

Probe Tumor Sentinel lymph nodes

FIG. 19-1  SLN biopsy for the treatment of breast cancer. A, Radioactive isotope and/or blue dye is injected into the tissue surrounding the tumor. B, The injected material migrates to draining lymph nodes. Sentinel nodes are identified visually or with a gamma-detecting probe. C, The tumor and sentinel nodes are excised for pathologic assessment of the regional lymph nodes.

excised for immediate evaluation; on identification of tumor cells in the SLN, a total lymph node dissection was done. At about this time, Morton et al3 investigated the various techniques of SLN identification—first in a feline model and subsequently in patients—by the intraoperative injection of isosulfan blue dye adjacent to primary melanoma sites to identify SLNs in the regional nodal basin3 (Fig. 19-1). In the early 1990s, SLNB was adopted for the pathologic staging of regional lymph nodes in patients with breast cancer. In 1996 a prospective trial of patients with invasive breast cancer who underwent SLNB before a completion axillary lymph node dissection (ALND) and segmental mastectomy or mastectomy within a single surgical procedure was reported.4 In this trial the SLN was identified in 92% of patients, and a positive SLN was identified before ALND in all patients with positive nodal disease. Albertini et al4 reported that this technique reduced surgical morbidity. This technique also gave pathologists the opportunity for an in-depth evaluation of fewer lymph nodes to more accurately identify micrometastatic disease. SLNB has also been applied to gynecologic cancers. This procedure was initially piloted for the treatment of squamous cell carcinoma of the vulva.5 In a prospective study of women undergoing routine lymphadenectomy, surgeons identified and excised the SLNs with preoperative lympho­ scintigraphy and intraoperative blue dye and subsequently performed a completion inguinofemoral lymph node dissection (ILND). Metastatic nodal disease was pathologically confirmed in almost half of the patients, each of whom had a positive SLN.5 Breast and cervical cancers are the first and fourth most commonly diagnosed cancers in women worldwide; an estimated 1,676,633 new cases of breast cancer and 1,085,948 new cases of cervical

K23349_Neligan_19_Sentinal Lymph_r5_dc_0263-0274.indd 264

5/27/2015 9:31:48 AM

265

Chapter 19  Sentinel Lymph Node Biopsy Outcomes

cancer are diagnosed annually.6 Melanoma is one of only a handful of cancers with an increasing annual incidence worldwide; an estimated 232,130 cases were diagnosed worldwide in 2012.6 With an overall estimated incidence of lymphedema of 6% for breast cancer, 4% for melanoma, and 9% for gynecologic cancers after SLNB, we estimate that 207,617 people will be diagnosed annually worldwide with lymphedema after SLNB, and many more are at a significant lifetime risk.7

Surgical Technique The current standard technique for SLNB is as follows. To facilitate the accurate identification of SLNs, blue dye is injected at the site of the primary tumor, turning the SLNs blue. Radioisotopes can also be injected at the primary tumor to guide surgeons to SLNs harboring radioactivity, regardless of color uptake (Fig. 19-2). Current techniques for SLNB most commonly use technetium99m (99mTc)-sulfur colloid (in the United States), 99mTc-nanocolloid (widely used in Europe), or

A

Sentinel lymph node

Melanoma

Afferent lymphatic channels

Afferent lymphatic channels

2 cm margin around lesion

B

Melanoma

FIG. 19-2  A, SLN biopsy for the evaluation of melanoma. Radioactive isotope and blue dye are injected into the tissue surrounding the lesion. Sentinel nodes are identified, exposed, and excised for pathologic assessment. B, Wide local excision for the removal of the primary tumor.

K23349_Neligan_19_Sentinal Lymph_r5_dc_0263-0274.indd 265

5/27/2015 9:31:49 AM

266

Part III  Pathophysiology and Clinical Presentation

A

B

FIG. 19-3  A, SLNB with a positive gross tumor. B, Positive tumor cells on pathologic analysis.

99mTc-antimony

trisulfide colloid (used in Australia) as a radiopharmaceutical agent.8 After the blue dye and radioactive isotope are injected, the surgeon uses a gamma-detecting probe to help localize sites of high radioactivity in the lymph node basin (see Fig. 19-2). An incision is made at a regional nodal basin site that has significant radioactivity, and the nodal tissue is dissected to identify blue and/or radioactive lymph nodes. The SLN or SLNs are excised from the remaining contents of the nodal basin and undergo pathologic assessment (Fig. 19-3). The accuracy and sensitivity of SLNB have been shown in most cancer types in which the procedure is part of the current treatment recommendations. When SLNB was done according to the standard treatment guidelines, the accuracy was 97%, sensitivity was 91%, specificity was 100%, and negative-predictive value was 95% in breast cancer.9 Similar numbers have been found in melanoma studies. A systematic review and meta-analysis by Meads et al10 evaluating SLNB in vulvar cancer found that the accuracy of the procedure (when done with both a blue dye and radioactive tracer) was 98%, sensitivity was 95%, and negative-predictive value was 98%.

Impact on Outcomes Long-Term Survival Outcomes The first evaluation of the impact of SLNB on survival was conducted in a multiinstitutional, randomized, controlled trial, the Multicenter Selective Lymphadenectomy Trial (MSLT-1), led by Morton.11 In this trial patients with melanoma were randomly assigned to receive a wide local excision and either SLNB or nodal observation only. Ten-year follow-up data on more than 1500 patients were available for the final analysis, which was published in 2014.12 Statistically, the 10year melanoma-specific survival rates were significantly higher in patients who received SLNB (81.4% 6 1.5%) than in those who had nodal observation only (78.3% 6 2.0%). In light of these findings, the National Comprehensive Cancer Network guidelines have included recommendations for the pathologic staging of melanoma in patients with primary tumors that are thicker than 0.75 mm or a primary tumor of any thickness that is ulcerated or has at least one or more mitotic figures per high-power field.13

K23349_Neligan_19_Sentinal Lymph_r5_dc_0263-0274.indd 266

5/27/2015 9:31:49 AM

Chapter 19  Sentinel Lymph Node Biopsy Outcomes

267

Lymphedema Outcomes Lymphedema has long been recognized as a potential morbidity after a complete lymph node dissection for many solid tumors.7 In a systematic review of non–breast cancer lymphedema, Cormier et al7 identified 47 studies across five categories of malignancies (melanoma, gynecologic cancer, genitourinary cancer, head and neck cancer, and sarcoma) that reported incidences of lymphedema ranging from 4% to 40% after complete lymph node dissection. Recently SLNB has also been recognized as a risk factor for lymphedema; however, the risk is significantly less than that of complete lymph node dissection. Tables 19-1, 19-2, and 19-3 summarize the peer-reviewed publications (2000 to 2014) reporting the incidence of lymphedema after SLNB for breast cancer, melanoma, and gynecologic malignancies. There is significant heterogeneity in the measurement techniques used to quantify lymphedema, which included water displacement, perometry, and limb circumference measurement.

Melanoma Six studies that included patients with melanoma undergoing SLNB and reported data on lymphedema outcomes were identified. The weighted pooled incidence of lymphedema was 4% (range 0.6% to 15%) (Table 19-1). In a systematic review of lymphedema in non–breast cancer solid tumors, Cormier et al7 found that the pooled lymphedema incidence was 18% after ILND and 3% after ALND. In a prospective study designed to assess lymphedema after SLNB or complete lymph node dissection (axillary or inguinofemoral) for the pathologic assessment and treatment of cutaneous melanoma, Hyngstrom et al14 used perometry to measure the limb circumference of patients with lymphedema before and after surgical intervention (lymphedema was defined as a 10% increase in limb volume from baseline) and assessed subjective, patient-reported symptoms of lymphedema with a validated instrument, the 19-item Lymphedema and Melanoma Questionnaire. Twelve months after surgery, 15% of patients who underwent SLNB and 30% of those who

TABLE 19-1  Incidence of Lymphedema 12 Months After SLNB for the Treatment of Melanoma Author (yr)

Number of Patients

Hyngstrom et al14 (2013) 15

Murawa et al (2013) 16

Measurement Technique

Incidence of Lymphedema (%)

84

Perometry

15

47

Circumference

2

Palmer et al (2013)*

47

Not reported

2

de Vries et al17 (2006)

52

Circumference

6

de Vries et al18 (2005)

44

Water displacement

11

Roaten et al (2005)

339

Not reported

0.6

Total studies (N 5 6)

613

19

*Pediatric melanoma patient cohort. Mean: 6.1 (range 0.6-15); weighted pooled incidence: 4.1.

K23349_Neligan_19_Sentinal Lymph_r5_dc_0263-0274.indd 267

5/27/2015 9:31:49 AM

268

Part III  Pathophysiology and Clinical Presentation

TABLE 19-2  Incidence of Lymphedema 12 Months After SLNB for the Treatment of Breast Cancer Author (yr)

Number of Patients

Sackey et al21 (2014) 22

Sagen et al (2014) 23

Measurement Technique

Incidence of Lymphedema (%)

140

Water displacement

20

187

Water displacement

3

Velloso et al (2011)

45

Circumference

4

Goldberg et al24 (2010)

600

Circumference

5

Lucci et al25 (2007)*

446

Circumference

7

26

449

Circumference

3.5

27

Mansel et al (2006)

478

Circumference

5

Francis et al28 (2006)

26

Circumference

17

2904

Circumference

7

Not reported

4

Langer et al (2007)

Wilke et al20 (2006) 29

Leidenius et al (2004) 30

92

Ronka et al (2004)

57

Not reported

23

Langer et al31 (2004)

40

Not reported

0

683

Circumference

6

Haid et al (2002)

57

Circumference

4

Swenson et al34 (2002)

169

Subjective

9

35

303

Not reported

3

Schrenk et al (2000)

35

Not reported

0

Total studies (N 5 17)

6711

32

Blanchard et al (2003) 33

Sener et al (2001) 36

*Chosen from multiple papers with overlapping patients. Mean: 7 (range 0-23); weighted pooled incidence: 6.3.

underwent lymph node dissections had objectively measured lymphedema. The highest incidence of lymphedema was in patients who had an ILND, 46% of whom had objectively measured lymphedema 12 months after surgery.14

Breast Cancer In the breast cancer literature, 17 studies (6711 patients) reporting on the incidence of lymphedema after SLNB were identified (Table 19-2). In these studies the weighted pooled incidence of breast cancer–related lymphedema in patients who underwent SLNB as their most invasive regional nodal procedure was 6% (range 0% to 23%). Wilke et al20 were one of the first groups to examine surgical complications after SLNB in patients with breast cancer. In a multiinstitutional trial more than 5000 patients were evaluated for lymphedema 30 days and 6 months after surgery. Lymphedema was measured with the use of limb circumference; the threshold for lymphedema was defined as a 2 cm difference between the affected and unaffected limbs. Lymphedema was observed in 7% of patients 6 months after surgery, and a higher body mass index was identified as a statistically significant risk factor for the development of lymphedema.

K23349_Neligan_19_Sentinal Lymph_r5_dc_0263-0274.indd 268

5/27/2015 9:31:49 AM

269

Chapter 19  Sentinel Lymph Node Biopsy Outcomes

TABLE 19-3  Incidence of Lymphedema 12 Months After SLNB for the Treatment of Gynecologic Cancer Author (yr)

Number of Patients

Measurement Technique

Incidence of Lymphedema (%)

Robison et al39 (2014)

69

Not reported

8

Achouri et al38 (2013)

88

Subjective

11

Novackova et al40 (2012)

12

Circumference

25

37

23

Subjective

9

Moore et al (2008)

31

Not reported

0

Total (N 5 5)

223

Niikura et al (2012) 41

Mean: 10.6 (range 0-25); weighted pooled incidence: 9.0.

In another study, Lucci et al25 prospectively compared postoperative complications in almost 900 patients who underwent either ALND or SLNB. In this study lymphedema was defined objectively as a 2 cm difference in circumference between the affected and unaffected arms and subjectively with patient self-reports. Using objective measurements, the 12-month lymphedema incidences were 11% in the ALND group and 6% in the SLNB group. Because of the relatively small sample size of patients who agreed to participate in the lymphedema measurement part of the study, this difference was not statistically significant. However, according to the subjective outcomes, 19% of patients who had ALND and 6% of patients who had SLNB had lymphedema, and these differences were statistically significant.25 The cumulative incidence of lymphedema over time was not reported in this study, which may explain the lower incidence reported for the ALND group compared with the historical lymphedema incidences in patients who had this procedure. Another potential explanation for the lower than expected reported incidence in the ALND group is that the circumference was measured at only one point on the arm, a method that is less sensitive to changes in limb size than multiple measurements per limb.

Gynecologic Cancers For gynecologic malignancies, five studies (223 patients who underwent SLNB) had a weighted pooled incidence of lymphedema of 9% (range 0% to 25%) (Table 19-3). These studies included patients with cervical,37,38 vulvar,38-40 and endometrial cancer.38 All of these studies were conducted after biopsy and lacked baseline measurements, which may have impacted the outcomes. Only Novackova et al40 used objective measurements of lymphedema (circumference).

Quality of Life Outcomes Because of improved treatment outcomes and earlier detection, more people have survived cancer in recent years than previously. Thus it is essential to evaluate how cancer treatment impacts QOL. With the widespread use of SLNB for the nodal assessment of many malignancies, QOL after SLNB is an important consideration.

K23349_Neligan_19_Sentinal Lymph_r5_dc_0263-0274.indd 269

5/27/2015 9:31:49 AM

270

Part III  Pathophysiology and Clinical Presentation

Health-related QOL is a multidimensional construct that includes not only physical well-being but also functional, emotional, and social domains.42 Physical well-being includes domains related to symptoms (for example, pain, discomfort, and fatigue), and functional well-being is the ability to complete day-to-day tasks (for example, bathing, eating, sleeping, and dressing). Emotional well-being captures a person’s ability to cope with various situations and to manage feelings ranging from distress and anxiety to enjoyment and happiness. Social well-being includes measures of the quality of relationships with family and friends and interactions with others in a wider social context.43

Melanoma Only the study by de Vries et al44 reported on the QOL of patients after SLNB for the treatment of melanoma. This study used the European Organization for Research and Treatment of Cancer QOL generic instrument (EORTC QLQ-C30) and a pain and activity questionnaire to assess 116 patients undergoing an axillary or inguinal SLNB or total lymphadenectomy. The study’s findings reported that patients undergoing SNLB statistically had significantly more favorable scores in physical and role functioning in the EORTC domains than those who underwent total lymphadenectomy, but no other statistically significant differences were identified between the two groups.

Breast Cancer QOL after SLNB for the treatment of breast cancer has been evaluated primarily with the use of two instruments: the EORTC QLQ-C30 with the associated breast cancer module (EORTC QLQ-BR23)45-47 and the Functional Assessment of Cancer Therapy Breast Cancer module (FACTB14).48 In addition, functional well-being after SLNB has been assessed with the Disabilities of the Arm, Shoulder, and Hand (DASH) instrument.49 A prospective, longitudinal study that evaluated women undergoing surgery for breast cancer assessed QOL with the EORTC QLQ-BR23 and EORTC QLQ-C30 before surgery and three times after surgical intervention with 1 year of total follow-up.47 Before surgery, patients had similar QOL regardless of expected surgery. Global health status declined in the immediate postoperative period but returned to preoperative levels 12 months after surgery; similar findings were observed with the breast and arm disability scales.47 Dabakuyo et al47 reported that patients who underwent SLNB alone and those who underwent ALND with or without SLNB did not significantly differ in global QOL or symptom-specific scales at 12 months. However, a large amount of QOL data were missing because of patient attrition, which may have biased the results. A comparison of QOL outcomes between patients undergoing ALND and those undergoing SLNB is important, considering the reduced morbidity associated with SLNB. A prospective longitudinal comparison study of patients undergoing either ALND or SLNB evaluated patients preoperatively and at three time points in the 12 months after surgery with the FACT-B14 questionnaire.48 Patients in the SLNB group reported deterioration from baseline in arm disability at both 6 and 12 months, but this statistically significant deterioration was much less than that of the ALND group. In a study of the long-term impact of SLNB on the QOL of breast cancer patients, De Gournay et al45 prospectively assessed more than 500 patients preoperatively and for as long as 72 months

K23349_Neligan_19_Sentinal Lymph_r5_dc_0263-0274.indd 270

5/27/2015 9:31:49 AM

Chapter 19  Sentinel Lymph Node Biopsy Outcomes

271

after surgery with the EORTC QLQ-BR23 and EORTC QLQ-C30 questionnaires. At 72 months after surgery, patients undergoing SLNB statistically had significantly better arm symptom scores than those who received ALND. In a subset analysis, patients who had a positive SLN and subsequently underwent ALND and those who underwent ALND only did not significantly differ in arm symptoms, breast symptoms, or global QOL at any time point. Global QOL scores were similar between the ALND and SLNB groups at baseline, declined in the immediate postoperative period, and returned to baseline level 12 months after surgery. Scores declined again at 12 months, although these scores did not significantly differ when stratified by surgical group.

Gynecologic Cancers QOL after SLNB has not been assessed as much for patients with gynecologic cancers as it has been in patients with breast cancer. Oonk et al50 used the EORTC QLQ-C30 and a subscale specific to vulvar cancer in an assessment of the efficacy of SLNB in vulvar cancer at various points in the course of care.50 The differences were assessed between the general QOL and symptom scale within the EORTC QLQ-C30 and additional symptom subscale; the only significant difference between the SLNB and ILND groups was that patients undergoing ILND reported greater financial difficulties. For the vulva-specific subscale, the statistically significant differences between the two groups included higher levels of contentment and lower levels of edema than those treated with ILND.50 In this evaluation patients were asked whether they would undergo a SLNB given a certain false-negative rate or an ILND at the initiation of treatment; 82% of the entire cohort (97% of the SLNB group and 62% of the ILND group) responded that they preferred the SLNB if the false-negative rate was no more than 1%, considering the reduced morbidity of SLNB.

Conclusion The introduction of SLNB across multiple solid tumor cancers has helped increase 5-year survival and reduced the morbidity associated with total lymphadenectomy. In addition to the improved survival outcomes, SLNB is a less invasive surgical procedure, which means less time under general anesthesia, a reduced risk to develop postoperative infection, and a better cosmetic outcome. Although SLNB significantly reduces the risk of postoperative lymphedema compared with completion lymph node dissection, it does not eliminate the risk entirely. Therefore when obtaining operative consent for SLNB, the surgeon must inform the patient that lymphedema is a possible postoperative long-term morbidity.

C linical P earls • SNL procedures were developed and validated in the 1990s for melanoma and breast cancer. • The incidence of lymphedema after SNLB ranges from 4% to 9% and avoids the risk of completion lymph node dissection for patients who are node negative. • Patient-reported QOL is affected less by SLNB than by total lymphadenectomy.

K23349_Neligan_19_Sentinal Lymph_r5_dc_0263-0274.indd 271

5/27/2015 9:31:49 AM

272

Part III  Pathophysiology and Clinical Presentation

R EFERENCES 1. Gould EA, Winship T, Philbin PH, et al. Observations on a “sentinel node” in cancer of the parotid. Cancer 13:77-78, 1960. 2. Cabanas RM. An approach for the treatment of penile carcinoma. Cancer 39:456-466, 1977. 3. Morton DL, Wen DR, Wong JH, et al. Technical details of intraoperative lymphatic mapping for early stage melanoma. Arch Surg 127:392-399, 1992. 4. Albertini JJ, Lyman GH, Cox C, et al. Lymphatic mapping and sentinel node biopsy in the patient with breast cancer. JAMA 276:1818-1822, 1996. 5. de Hullu JA, Hollema H, Piers DA, et al. Sentinel lymph node procedure is highly accurate in squamous cell carcinoma of the vulva. J Clin Oncol 18:2811-2816, 2000. 6. World Health Organization. GLOBOCAN 2012: Estimated Cancer Incidence, Mortality and Prevalence Worldwide in 2012. Lyon, France: International Agency for Research on Cancer, 2012. 7. Cormier JN, Askew RL, Mungovan KS, et al. Lymphedema beyond breast cancer: a systematic review and meta-analysis of cancer-related secondary lymphedema. Cancer 116:5138-5149, 2010. 8. Thompson JF, Morton DL. Lymphatic mapping and sentinel lymph node biopsy: the concept. In Thompson JF, Morton DL, Kroon BBR, eds. Textbook of Melanoma. London: Taylor & Francis, 2004. 9. Veronesi U, Paganelli G, Viale G, et al. A randomized comparison of sentinel-node biopsy with routine axillary dissection in breast cancer. N Engl J Med 349:546-553, 2003. 10. Meads C, Sutton AJ, Rosenthal AN, et al. Sentinel lymph node biopsy in vulval cancer: systematic review and meta-analysis. Br J Cancer 110:2837-2846, 2014. 11. Morton DL, Cochran AJ, Thompson JF, et al; Multicenter Selective Lymphadenectomy Trial Group. Sentinel node biopsy for early-stage melanoma. Accuracy and morbity in MSLT-1, an International Multicenter Trial. Ann Surg 242:302-313, 2005. 12. Morton DL, Thompson JF, Cochran AJ, et al. Final trial report of sentinel-node biopsy versus nodal observation in melanoma. N Engl J Med 370:599-609, 2014. 13. NCCN Guidelines for Physicians: Melanoma. Fort Washington, PA: National Comprehensive Cancer Network, 2014. 14. Hyngstrom JR, Chiang YJ, Cromwell KD, et al. Prospective assessment of lymphedema incidence and lymphedema-associated symptoms following lymph node surgery for melanoma. Melanoma Res 23:290-297, 2013. 15. Murawa D, Polom K, Murawa P. One-year postoperative morbidity associated with near-infraredguided indocyanine green (ICG) or ICG in conjugation with human serum albumin (ICG:HSA) sentinel lymph node biopsy. Surg Innov 21:240-243, 2013. 16. Palmer PE III, Warneke CL, Hayes-Jordan AA, et al. Complications in the surgical treatment of pediatric melanoma. J Pediatr Surg 48:1249-1253, 2013. 17. de Vries M, Vonkeman WG, van Ginkel RJ, et al. Morbidity after inguinal sentinel lymph node biopsy and completion lymph node dissection in patients with cutaneous melanoma. Eur J Surg Oncol 32:785789, 2006. 18. de Vries M, Vonkeman WG, van Ginkel RJ, et al. Morbidity after axillary sentinel lymph node biopsy in patients with cutaneous melanoma. Eur J Surg Oncol 31:778-783, 2005. 19. Roaten JB, Pearlman N, Gonzalez R, et al. Identifying risk factors for complications following sentinel lymph node biopsy for melanoma. Arch Surg 140:85-89, 2005. 20. Wilke LG, McCall LM, Posther KE, et al. Surgical complications associated with sentinel lymph node biopsy: results from a prospective international cooperative group trial. Ann Surg Oncol 13:491-500, 2006. 21. Sackey H, Magnuson A, Sandelin K, et al. Arm lymphoedema after axillary surgery in women with invasive breast cancer. Br J Surg 101:390-397, 2014. 22. Sagen A, Kaaresen R, Sandvik L, et al. Upper limb physical function and adverse effects after breast cancer surgery: a prospective 2.5-year follow-up study and preoperative measures. Arch Phys Med Rehabil 95:875-881, 2014.

K23349_Neligan_19_Sentinal Lymph_r5_dc_0263-0274.indd 272

5/27/2015 9:31:49 AM

Chapter 19  Sentinel Lymph Node Biopsy Outcomes

273

23. Velloso FS, Barra AA, Dias RC. Functional performance of upper limb and quality of life after sentinel lymph node biopsy of breast cancer. Rev Bras Fisioter 15:146-153, 2011. 24. Goldberg JI, Wiechmann LI, Riedel ER, et al. Morbidity of sentinel node biopsy in breast cancer: the relationship between the number of excised lymph nodes and lymphedema. Ann Surg Oncol 17:32783286, 2010. 25. Lucci A, McCall LM, Beitsch PD, et al. Surgical complications associated with sentinel lymph node dissection (SLND) plus axillary lymph node dissection compared with SLND alone in the American College of Surgeons Oncology Group Trial Z0011. J Clin Oncol 25:3657-3663, 2007. 26. Langer I, Guller U, Berclaz G, et al. Morbidity of sentinel lymph node biopsy (SLN) alone versus SLN and completion axillary lymph node dissection after breast cancer surgery: a prospective Swiss multicenter study on 659 patients. Ann Surg 245:452-461, 2007. 27. Mansel RE, Fallowfield L, Kissin M, et al. Randomized multicenter trial of sentinel node biopsy versus standard axillary treatment in operable breast cancer: the ALMANAC Trial. J Natl Cancer Inst 98:599609, 2006. 28. Francis WP, Abghari P, Du W, et al. Improving surgical outcomes: standardizing the reporting of incidence and severity of acute lymphedema after sentinel lymph node biopsy and axillary lymph node dissection. Am J Surg 192:636-639, 2006. 29. Leidenius M, Krogerus L, Tukiainen E, et al. Accuracy of axillary staging using sentinel node biopsy or diagnostic axillary lymph node dissection—a case-control study. APMIS 112:264-270, 2004. 30. Ronka R, Smitten K, Sintonen H, et al. The impact of sentinel node biopsy and axillary staging strategy on hospital costs. Ann Oncol 15:88-94, 2004. 31. Langer S, Guenther JM, Haigh PI, et al. Lymphatic mapping improves staging and reduces morbidity in women undergoing total mastectomy for breast carcinoma. Am Surg 70:881-885, 2004. 32. Blanchard E, Herman L, Larson L, et al. Understanding the role of sentinel lymph node biopsy in breast cancer and melanoma. JAAPA 16:49-50, 54, 2003. 33. Haid A, Koberle-Wuhrer R, Knauer M, et al. Morbidity of breast cancer patients following complete axillary dissection or sentinel node biopsy only: a comparative evaluation. Breast Cancer Res Treat 73:31-36, 2002. 34. Swenson KK, Nissen MJ, Ceronsky C, et al. Comparison of side effects between sentinel lymph node and axillary lymph node dissection for breast cancer. Ann Surg Oncol 9:745-753, 2002. 35. Sener SF, Winchester DJ, Martz CH, et al. Lymphedema after sentinel lymphadenectomy for breast carcinoma. Cancer 92:748-752, 2001. 36. Schrenk P, Rieger R, Shamiyeh A, et al. Morbidity following sentinel lymph node biopsy versus axillary lymph node dissection for patients with breast carcinoma. Cancer 88:608-614, 2000. 37. Niikura H, Okamoto S, Otsuki T, et al. Prospective study of sentinel lymph node biopsy without further pelvic lymphadenectomy in patients with sentinel lymph node-negative cervical cancer. Int J Gynecol Cancer 22:1244-1250, 2012. 38. Achouri A, Huchon C, Bats AS, et al. Complications of lymphadenectomy for gynecologic cancer. Eur J Surg Oncol 39:81-86, 2013. 39. Robison K, Roque D, McCourt C, et al. Long-term follow-up of vulvar cancer patients evaluated with sentinel lymph node biopsy alone. Gynecol Oncol 133:416-420, 2014. 40. Novackova M, Halaska MJ, Robova H, et al. A prospective study in detection of lower-limb lymphedema and evaluation of quality of life after vulvar cancer surgery. Int J Gynecol Cancer 22:1081-1088, 2012. 41. Moore RG, Robison K, Brown AK, et al. Isolated sentinel lymph node dissection with conservative management in patients with squamous cell carcinoma of the vulva: a prospective trial. Gynecol Oncol 109:65-70, 2008. 42. Cella DF, Tulsky DS, Gray G, et al. The Functional Assessment of Cancer Therapy scale: development and validation of the general measure. J Clin Oncol 11:570-579, 1993. 43. Cella D, Nowinski CJ. Measuring quality of life in chronic illness: the functional assessment of chronic illness therapy measurement system. Arch Phys Med Rehabil 83(12 Suppl 2):S10-S17, 2002.

K23349_Neligan_19_Sentinal Lymph_r5_dc_0263-0274.indd 273

5/27/2015 9:31:49 AM

274

Part III  Pathophysiology and Clinical Presentation

44. de Vries M, Hoekstra HJ, Hoekstra-Weebers JE. Quality of life after axillary or groin sentinel lymph node biopsy, with or without completion lymph node dissection, in patients with cutaneous melanoma. Ann Surg Oncol 16:2840-2847, 2009. 45. De Gournay E, Guyomard A, Coutant C, et al. Impact of sentinel node biopsy on long-term quality of life in breast cancer patients. Br J Cancer 109:2783-2791, 2013. 46. Boguševičius A, Čepulienė D. Quality of life after sentinel lymph node biopsy versus complete axillary lymph node dissection in early breast cancer: a 3-year follow-up study. Medicina (Kaunas) 49:111-117, 2013. 47. Dabakuyo TS, Fraisse J, Causeret S, et al. A multicenter cohort study to compare quality of life in breast cancer patients according to sentinel lymph node biopsy or axillary lymph node dissection. Ann Oncol 20:1352-1361, 2009. 48. Belmonte R, Garin O, Segura M, et al. Quality-of-life impact of sentinel lymph node biopsy versus axillary lymph node dissection in breast cancer patients. Value Health 15:907-915, 2012. 49. Hudak PL, Amadio PC, Bombardier C. Development of an upper extremity outcome measure: the DASH (disabilities of the arm, shoulder and hand) [corrected]. The Upper Extremity Collaborative Group (UECG). Am J Ind Med 29:602-608, 1996. 50. Oonk MH, van Os MA, de Bock GH, et al. A comparison of quality of life between vulvar cancer patients after sentinel lymph node procedure only and inguinofemoral lymphadenectomy. Gynecol Oncol 113:301-305, 2009.

K23349_Neligan_19_Sentinal Lymph_r5_dc_0263-0274.indd 274

5/27/2015 9:31:50 AM

Part IV

Diagnosis of Lymphedema

K23346_Neligan_20_Causes_r3_0275-0290.indd 275

5/26/2015 9:21:59 AM

K23346_Neligan_20_Causes_r3_0275-0290.indd 276

5/26/2015 9:22:00 AM

C hapter 20 Causes and Classification of Lymphatic Disorders Swetha Kambhampati, Stanley Rockson

K ey P oints • Primary lymphedema is generally categorized based on age of onset and is presumed to have a genetic basis. • Lymphedema is a hallmark feature of various syndromic disorders. • Secondary lymphedema arises as a consequence of other conditions or treatments. • Any surgical intervention can precipitate lymphedema. • Lymphangiomatosis can be isolated or systemic, superficial or visceral. It is generally characterized by the depth of the malformation within the tissue planes. • Lymphatic filariasis results from an infection and worldwide is the most common cause of lymphedema. • Abnormal development or damage to blood and lymphatic vascular systems can contribute to the appearance of complex vascular malformations.

Cau

• Protein-losing enteropathy and intestinal lymphangiectasia are associated with the loss of lymphatic fluid and plasma protein within the lumen of the gastrointestinal tract.

Lymphedema is a complex edematous state resulting from dysfunction of the lymphatic transport system that is essential to the maintenance of tissue homeostasis. Beyond lymphedema, however, there is a broad spectrum of diseases that directly or indirectly alter lymphatic structure and function. These lymphatic vascular diseases, which are characterized by a failure of adequate lymph transport, lead to a wide range of pathologic presentations.1 Given the central role of lymphatic function in circulatory homeostasis and a robust immune and metabolic contribution (based on suitable dietary absorption of gastrointestinal lipids), any lymphatic pathology will become clinically manifest as a regional or systemic immune impairment and/or significant metabolic derangement, often accompanied by chronic and often debilitating regional swelling and tissue edema.2-4

277

K23346_Neligan_20_Causes_r3_0275-0290.indd 277

5/26/2015 9:22:00 AM

278

Part IV  Diagnosis of Lymphedema

Embryologic Development The lymphatic system arises from endothelial cells within the embryonic venous structures. Specific molecular markers determine the lymphatic endothelial cell specification and thus its unique phenotype. Additional lymphatic-specific markers are expressed, and blood vascular expression profiles are suppressed as the lymphatic endothelial cells continue to differentiate.5 Vascular endothelial growth factor C is necessary for the earliest steps of lymphatic endothelial cell differentiation.6 The differentiated lymphatic cell population migrates peripherally and establishes complete autonomy from the local venous environment. This developmental stage is characterized by primary budding. The primary lymph sacs form throughout the embryo, followed by secondary budding and migration. The latter requires endothelial sprouting into tissues to form local capillaries, thereby marking the final stages of lymphatic development.7,8

Lymphedema The International Society of Lymphology consensus document has identified four stages in the natural history of lymphedema.9 Stage 0 refers to a latent or subclinical condition in which swelling is not seen despite the presence of impairment in lymph transport. Stage 1 is seen as pitting edema. There is early accumulation of fluid that is relatively high in protein content and resolves with limb elevation. Stage 2 is characterized by the presence of pitting edema that does not resolve with limb elevation alone. Stage 3 is lymphostatic elephantiasis, in which pitting is no longer seen. The lymphedema is characterized by trophic skin changes, such as acanthosis, fat deposition, dermal cutaneous fibrosis, and verrucous overgrowth.10 Although lymphedema can be hereditary, its development can also be sporadic. Its cause is either primary or secondary: • Primary lymphedemas are congenital, developmental, or inherited disorders that arise from an inborn malformation or dysfunction of the lymphatic system. • Secondary lymphedema arises as a consequence of other conditions or treatments. Lymphedema is not a reversible condition. Its treatment is largely mechanical and requires physiotherapeutic interventions that reduce limb swelling through the stimulation of lymphatic contractility and that control reaccumulation of edema through limb compression. Complete decongestive therapy is a multicomponent treatment that reduces the degree of lymphedema and maintains skin health.11-13 At present there is no long-term pharmacologic therapy that is recommended for patients with lymphedema.

Primary Lymphedema Primary lymphedemas are congenital, developmental, or inherited disorders that arise from an inborn malformation or dysfunction of the lymphatic system. In general, these disorders do not shorten life expectancy but worsen quality of life.14 They are presumed to have a genetic basis; an

K23346_Neligan_20_Causes_r3_0275-0290.indd 278

5/26/2015 9:22:00 AM

Chapter 20  Causes and Classification of Lymphatic Disorders

279

autosomal dominant transmission pattern is most often described. Numerous distinct gene mutations associated with lymphedemas have thus far been noted and characterized.15,16 Primary lymphedemas are classified by the age of onset: • Congenital lymphedema presents at birth or by the age of 2 years • Lymphedema praecox presents before 35 years of age (most often in association with puberty). • Lymphedema tarda appears after 35 years of age. Congenital lymphedema can be syndromic in nature (for example, Noonan syndrome, Turner syndrome, or Klippel-Trenaunay syndrome)16,17 and can be organized by its pattern of inheritance. Chromosomal disorders, such as Klinefelter or Edwards syndrome, can result in multiple organ defects and a wide constellation of symptoms and complications, including a severe distortion of lymphatic function.3 Typically, congenital lymphedema of the hands and feet is seen in infants with Turner syndrome.18 Milroy disease, or hereditary autosomal dominant congenital lymphedema, presents as painless, nonprogressive edema of the lower extremity in infancy.19 It has been associated with mutations in the kinase domain of vascular endothelial growth factor receptor 3 (VEGFR-3), with incomplete penetrance.20 Despite the association of Milroy disease with a systemic mutation, lymphoscintigraphy often reveals normal, functional lymphatic anatomy in the upper limbs, but inadequate lymphatic functional integrity is present at the sites of swelling in the lower limbs.19 Lymphedema-distichiasis is a single-gene disorder caused by a mutation in the gene that encodes the nuclear transcription factor FOXC2. This is another common cause of primary lymphedema. Distichiasis (abnormal cilia arising from the meibomian glands along the lid margin) typically presents at birth, but the lymphedema generally presents at puberty (often earlier in males)21; it is an accompanying somatic anomaly that is found in 94% to 100% of patients with lymphedemadistichiasis. This feature can lead to recurrent corneal irritation and conjunctivitis.19 FOXC2 mutations can also be associated with the presence of cleft palate or congenital heart disease. Lymphoscintigraphy reveals reflux within the main collector lymphatics; in addition, deep vein reflux is seen on venous Doppler ultrasound examination.19,22 Lymphedema praecox (lymphedema presenting before the age of 35) is the most common form of primary lymphedema.23 The edema in this disease is often unilateral and commonly limited to the foot and calf.24 The disorder is characterized by an initial presentation at puberty and female preponderance, leading to the hypothesis that estrogen may play a role in the expression of the lymphedema.24 Meige disease is a term that has been applied to a familial form of lymphedema praecox with pubertal onset and an autosomal dominant pattern of inheritance.19

Secondary Lymphedema Secondary lymphedema arises as a consequence of other conditions or treatments. However, there is a growing conviction that genetic factors contribute to the susceptibility to the stressors that elicit an ostensibly pure acquired lymphedema. In other words, in many cases, there may be a primary predisposition to the development of secondary lymphedema.

K23346_Neligan_20_Causes_r3_0275-0290.indd 279

5/26/2015 9:22:00 AM

280

Part IV  Diagnosis of Lymphedema

A common cause of acquired lymphedema is iatrogenic, as a result of treatment of malignancies. Furthermore, and less commonly, tumors can primarily obstruct lymphatic channels or infiltrate the lymphatic system, leading to lymphedema through obstruction of lymph flow. Treatment of a wide variety of cancers is associated with a significant incidence of lymphedema, including breast cancer, malignant melanoma, urogenital malignancies, lymphoma, and soft tissue sarcomas.25 Any surgery can result in lymphedema. The operative intervention can be for cancer (for example, a modified radical mastectomy or even a partial excision) or a non-cancer-related procedure, such as varicose vein ligation or saphenous vein harvesting for aortocoronary bypass.26,27 Trauma, infection, and burns can also lead to the development of lymphedema. Moreover, peripheral arterial disease can result in secondary lymphedema.26 Iatrogenic causes, such as intrathecal pump insertion or sirolimus administration, are common precipitators of lymphedema.28,29 Globally, infection is considered the leading cause of lymphedema. In tropical regions, endemic lymphatic filariasis predominates and is the most common worldwide cause of lymphedema.30

Acquired Conditions Infectious Diseases Lymphatic filariasis and lymphangitis are two conditions in which invading pathogens infect and infiltrate the lymphatic system, which leads to lymphatic dysfunction. The consequence of such events can often result in a chronic obstruction to lymph flow (acquired lymphedema), which is accompanied by the resulting impairment in regional immune function. More than 140 million people worldwide have been infected with filariasis. The prevalence and global burden of the disease lag only behind malaria and tuberculosis.31 Most of these infections are caused by Wuchereria bancrofti, whereas the remaining cases are caused by Brugia malayi.32 Patients are infected by the larvae of these filariae, or parasitic worms; after the larvae are delivered through the wound, they enter the draining lymphatic vessels. The larvae develop into mature adult worms that produce microfilariae, which then enter and circulate in the bloodstream. Lymphatic filariasis can be asymptomatic (subclinical) or can have acute or chronic clinical manifestations. Acute presentations include adenolymphangitis, which is characterized by fever and painful lymphadenopathy. Ultimately, there is fibrosis of the lymph nodes and impaired regeneration of lymphatic channels. Dermatolymphangioadenitis is another acute presentation that is characterized by inflammatory plaques and systemic symptoms as a consequence of superficial bacterial infection of damaged skin.33 Other acute presentations include filarial fevers and tropical pulmonary eosinophilia, which is caused by an immune response to filariae trapped in the lungs. Chronically, filariasis can lead to lymphedema, hydrocele, and renal involvement.34 Active infection can be diagnosed by the detection of the microfilariae in the blood; detection of localized, lymphatic obstruction by Doppler ultrasound is feasible.31 Serologic techniques can be substituted for the microscopic detection of microfilariae. The infection is treated with antibiotics and antiparasitic medications, such as albendazole, diethylcarbamazine, and ivermectin.35-37 Lymphangitis represents an inflammation of the lymphatic channels, most often seen in relation to infectious causes, including bacterial, mycobacterial, fungal, viral, and parasitic pathogens.38

K23346_Neligan_20_Causes_r3_0275-0290.indd 280

5/26/2015 9:22:00 AM

Chapter 20  Causes and Classification of Lymphatic Disorders

281

Lymphangitis often develops after cutaneous inoculation of microorganisms through a skin wound or as a complication of a distal infection that invades the lymphatic vessels and spreads toward regional lymph nodes. Lymphangitis can also reflect inflammation in the setting of malignancy (neoplastic lymphangitis or lymphangitis carcinomatosa) or a systemic inflammatory process, such as Crohn’s disease (granulomatous intestinal lymphangitis) or sclerosing lymphangitis of the penis.39-43 Recurrent episodes of bacterial lymphangitis lead to thrombosis and fibrosis of the lymphatic channels; this is one of the most common causes of lymphedema.44 The clinical presentation of this disease often encompasses the presence of painful erythematous cutaneous streaks and enlarged and tender lymph nodes.38 Patients also frequently have a history of minor trauma or skin infection and fever, chills, muscular pain, and headache.45 Imaging is rarely used for infectious causes, but lymphangiography and lymphoscintigraphy can be used to detect anatomic abnormalities, such as lymphatic obstruction or dilated, tortuous lymphatic vessels.46 Treatment most often involves the use of antimicrobial agents; some patients require surgical debridement.38,47

Congenital Conditions Lipedema Lipedema is a chronic disorder of adipose biology that results in symmetrical, bilateral, fatty subcutaneous tissue deposition in the lower extremities and buttocks. The condition is characterized by hyperplasia and hypertrophy of adipocytes. Lipedema can occur with lymphedema, although lipedema has distinctive features that allow it to be differentiated from the more common problem of lower extremity lymphedema. In lipedema, the skin is often sensitive to pressure. Affected individuals have both spontaneous pain and pressure-induced discomfort. The problem is also associated with easy bruising and hematoma development after even minimal trauma. In lipedema the Stemmer sign is absent, and the condition is described as foot sparing. Moreover, it is often characterized by a normal cutaneous architecture, with no demonstrable dermal fibrosis or cutaneous thickening. Lipedema almost exclusively affects females and is often associated with a familial distribution. Lipedema can ultimately cause secondary lymphedema (so-called lipolymphedema) by distorting the microlymphatic function.7,48-50 Therapies are mostly conservative and similar to those used in lymphedema, including complex decongestive therapy, pneumatic compression, manual lymphatic drainage, bandaging, and diet modifications.51,52

Lymphangiomatosis Lymphangiomatosis is a congenital lymphatic malformation that arises from abnormal embryologic development. When present, it is typically detected within the first 2 years of life. It is postulated to develop from abnormal grouping of lymph sacs during development or the failure of proper lymph vascular tissue anastomoses.53 There are three types of lymphangiomas, which are differentiated by the depth of the malformation54: 1. Lymphangioma circumscriptum (the most superficial of the three) 2. Cavernous lymphangiomas (characterized by deeper lesions) 3. Cystic hygromas (characterized by deeper lesions)

K23346_Neligan_20_Causes_r3_0275-0290.indd 281

5/26/2015 9:22:00 AM

282

Part IV  Diagnosis of Lymphedema

However, this terminology is in transition. The most useful diagnostic approach to lymphangioma is to perform an MRI, which yields anatomic information within the various tissue strata. Lymphangioma circumscriptum is distinguished by superficial, 1 to 2 mm vesicles in the papillary dermis. These lesions, which are filled with clear, colorless fluid, connect by dilated lymphatic channels to the subcutaneous lymphatic sacs.55,56 Cavernous lymphangiomas are microcystic soft tissue masses with lymphatic dilation in various layers beneath the epidermis of the tissue, including the dermis, subcutaneous tissue, and intermuscular septa.53 The overlying skin may show some hyperpigmentation and hyperplasia but is otherwise generally unaffected. Cystic hygromas are fluid-filled lesions caused by an obstruction in the drainage of the lymphatic system into the jugular vein, thus leading to the local accumulation of lymphatic fluid.53 These macrocystic fluid-filled sacs, which are often encased in a fibrous capsule, enlarge and fill dilated lymphatic vessels and tissue, leading to lymphedema.57 Cystic hygromas are associated with chromosomal disorders, such as Turner and Klinefelter syndromes.53 Lymphangiomatosis can present either as isolated lesions or as a systemic, multifocal derangement in lymphatic development. In the latter instance, in which visceral involvement is present, there is often significant pathologic dysfunction of the affected respiratory, gastrointestinal, or other organs. The treatment of lymphangioma may include surgical excision or chemical sclerosis.

Conditions With Congenital or Acquired Etiologic Factors Complex Vascular Malformations Complex vascular malformations manifest as a consequence of abnormal development or damage to blood and lymphatic vascular systems.16,58 These diseases often present with dermal manifestations, such as nodules or lesions. Cystic angiomatosis is a rare congenital condition characterized by multifocal hemangiomatous and/or lymphangiomatous lesions of the skeleton with possible visceral organ development.59 These cystic skeletal lesions may be comprised of dilated blood vessels, lymphatic channels, or both, and appear round or oval with a wide variety in size.60,61 Patients present with soft tissue masses and pathologic fractures that cause pain and swelling.61-63 On radiographs they appear as areas of destroyed bone encircled by a sclerotic rim and are confirmed by biopsy.60,61 Chemotherapy and radiotherapy have been tried, but generally they are ineffective.60 Maffucci syndrome is another type of complex vascular formation that is characterized by multiple enchondromas and subcutaneous hemangiomas resulting from mesodermal dysplasia.64-66 These tumors present early in childhood and progressively worsen, leading to significant skeletal deformities.64,66 Most of these tumors are benign, but there is a 15% to 30% incidence of malignant

K23346_Neligan_20_Causes_r3_0275-0290.indd 282

5/26/2015 9:22:00 AM

Chapter 20  Causes and Classification of Lymphatic Disorders

283

transformation, the most common of which is chondrosarcoma.64,65 This syndrome can lead to infectious complications and lymphedema.67 Diffuse hemangiomatosis is characterized by benign, visceral hemangiomas that affect at least three organ systems. The vascular hamartomas are a congenital defect and lead to diffuse lesions of the skin, liver, brain, lungs, and gastrointestinal tract. These lesions have a favorable response to corticosteroids and interferon-alfa therapy. Gorham disease is a rare condition characterized by the uncontrolled growth of benign blood and lymphatic vascular channels and the progressive growth of lymphangiomatous tissue that leads to massive osteolysis and bone destruction.68,69 This condition is also associated with chylous pericardial and pleural effusions.69 Surgical intervention (both resection or bone reconstruction) and radiotherapy are the treatment methods of choice.69-72 Proteus syndrome is a rare congenital hamartomatous disease characterized by the overgrowth of numerous body tissues and cell lines.73,74 It presents as subcutaneous tumors, hyperostosis, hyperplastic connective tissue in the palms and soles, pigmented nevi, and partial gigantism of the hands or feet.74,75 Klippel-Trenaunay syndrome (KTS) is a complex vascular malformation that includes capillary anomalies (port-wine stain), venous malformation (varicose veins), lymphatic anomalies, and hypertrophy of bone and soft tissue that can affect one or multiple limbs.17,76 The histologic hallmark of this condition is dilated telangiectatic vessels in the upper dermis that do not spontaneously regress.77 Complications of KTS include deep vein thrombosis, pulmonary embolism, gastrointestinal bleeding, and vascular (often lymphatic) blebs within the capillary malformations.78 Doppler ultrasonography can distinguish a KTS lesion from a hemangioma, but MRI and MR venography are useful to evaluate the extent of venous and lymphatic malformation and soft tissue and bone hypertrophy. Lymphoscintigraphy is useful for assessing the extent of lymphatic involvement.79-82 Treatment is focused on symptomatic relief; elevation and compression stockings offer some relief from the edema.83,84 Blue rubber bleb nevus is a rare, congenital venous malformation that consists of vascular nevi on the skin and hemangiomas of the gastrointestinal tract and soft tissues.85 The condition is characterized by anemia, fatigue, gastrointestinal bleeding, and cutaneous lesions that are painless, compressible, deep blue, and rubbery; the blebs are distributed diffusely.86 Treatment includes supportive medical therapy, such as transfusions and iron replacement for any gastrointestinal blood loss.85 Pharmacologic therapy, such as corticosteroids or interferon-alfa, have not been found to be very effective.87 Endoscopic interventions, such as sclerotherapy, can also be used to help control gastrointestinal blood loss.

Protein-Losing Enteropathy and Intestinal Lymphangiectasia Protein-losing enteropathy and intestinal lymphangiectasia are two disorders characterized by edema and hypoproteinemia and associated with the loss of lymphatic fluid and plasma protein within the lumen of the gastrointestinal tract. Although patients with protein-losing enteropathy have local lymphatic obstruction and stasis, those with lymphangiectasia have dilated lymphatic vessels in the intestinal villi.88,89

K23346_Neligan_20_Causes_r3_0275-0290.indd 283

5/26/2015 9:22:00 AM

284

Part IV  Diagnosis of Lymphedema

Protein-losing gastroenteropathies are characterized by an excessive loss of serum proteins into the gastrointestinal tract through the intestinal microvilli, resulting in hypoproteinemia, edema, and pleural and pericardial effusions. Protein-losing enteropathy is associated with various disorders, including inflammatory bowel disease, infection, rheumatic diseases, celiac disease, graft-versushost disease, cardiac disease, and primary intestinal lymphangiectasia.90 Protein loss in this type of disorder is nonselective, which is in contrast to the glomerular diseases.91 The clinical manifestations of protein-losing gastroenteropathies are highly variable and include edema, ascites, pleural and pericardial effusions, fat and carbohydrate malabsorption, and vitamin deficiencies. The diagnostic characteristics of this disorder include reduced serum concentrations of albumin, gamma globulins, fibrinogen, transferrin, and ceruloplasmin.90 The treatment of this condition largely depends on the underlying cause. Intravenous albumin replacement, small bowel resection, or high-dose steroid therapy may be beneficial.92-95 Severe reduction of dietary fat intake with supplementation of medium-chain triglycerides can be attempted. In patients with congenital cardiac disease, heparin administration is recommended.58,92 Primary intestinal lymphangiectasia is characterized by ectasia of enteral lymphatics, which are often located in the mucosa, submucosa, or subserosa. Clinical manifestations include severe edema, thickening of the small bowel wall, protein-losing enteropathy, chylothorax or chylous ascites, intermittent diarrhea, nausea, and vomiting. CT imaging can be used in the diagnosis of this condition and shows nodular thickening of the bowel wall with hypodense streaks in the small bowel, reflecting dilated lymphatics.96,97 Contrast lymphangiography, nuclear scintigraphy, and MR lymphangiography can also show abnormal intestinal lymphatics.98-100 Dietary fat restriction with medium-chain triglyceride supplementation has been found to be effective in this condition.101 Chylous ascites can be ameliorated through the administration of somatostatin or its analogs.102

Lymphangioleiomyomatosis Lymphangioleiomyomatosis is a disorder characterized by the cystic destruction of the lung and lymphatic wall thickening resulting from the migration of abnormal smooth muscle cells through the axillary lymphatics and pulmonary interstitium.103 Lymphangioleiomyomatosis is associated with pulmonary cysts and angiomyolipomas, tumors consisting of smooth muscle cells, adipose tissue, and underdeveloped blood vessels.104 The most common clinical presentation is pulmonary, with manifestations that include dyspnea, chylous pleural effusions, cough, hemoptysis, pulmonary hypertension, and pneumothorax.103 Chylous ascites is also frequently encountered. Nonpulmonary findings include lymphangioleiomyomas—benign neoplasms that present as cystic masses in the mediastinum, retroperitoneum, or pelvis.105,106 Renal angiomyolipomas and meningiomas are also associated with lymphangioleiomyomatosis.107-110 Tissue biopsy is the main diagnostic tool in lymphangioleimyomatosis.111 Treatment involves embolization and prevention of pneumothorax through pleurodesis and pleurectomy.58,112,113

K23346_Neligan_20_Causes_r3_0275-0290.indd 284

5/26/2015 9:22:00 AM

Chapter 20  Causes and Classification of Lymphatic Disorders

285

C linical P earls • In primary lymphedema, numerous distinct gene mutations have thus far been described and characterized. Clinical genetic testing is now available. • Trauma, infection, and iatrogenic factors predominate as the causes of acquired lymphedema. • Lipedema is a defect in the adipose biology that can be confused with lower extremity lymphedema. • Lymphatic and complex vascular malformations can be associated with the presence of superficial or visceral lymphedema. • Protein-losing enteropathy and intestinal lymphangiectasia are associated with the loss of lymphatic fluid and plasma protein within the lumen of the gastrointestinal tract. • Lymphangioleiomyomatosis is a rare, complex disorder that has a component of abnormal lymphatic biology and may encompass chylothorax or chylous ascites within its presentation.

R EFERENCES 1. Rockson SG. Physiology, pathophysiology, and lymphodynamics: general overview. In Lee BB, Bergan J, Rockson SG, eds. Lymphedema: A Concise Compendium of Theory and Practice. London: Springer-Verlag, 2011. 2. Ristevski B, Becker H, Cybulsky M, et al. Lymph, lymphocytes, and lymphatics. Immunol Res 35:5564, 2006. 3. Rockson SG. Lymphedema. Am J Med 110:288-295, 2001. 4. Harvey NL, Srinivasan RS, Dillard ME, et al. Lymphatic vascular defects promoted by Prox1 haploinsufficiency cause adult-onset obesity. Nat Genet 37:1072-1081, 2005. 5. Wigle JT, Harvey N, Detmar M, et al. An essential role for Prox1 in the induction of the lymphatic endothelial cell phenotype. EMBO J 21:1505-1513, 2002. 6. Karkkainen MJ, Haiko P, Sainio K, et al. Vascular endothelial growth factor C is required for sprouting of the first lymphatic vessels from embryonic veins. Nat Immunol 5:74-80, 2004. 7. Rockson SG. Etiology and classification of lymphatic disorders. In Lee BB, Bergan J, Rockson SG, eds. Lymphedema: A Concise Compendium of Theory and Practice. London: Springer-Verlag, 2011. 8. Oliver G, Harvey N. A stepwise model of the development of lymphatic vasculature. Ann N Y Acad Sci 979:159-165; discussion 188-196, 2002. 9. Lu P. Staging and classification of lymphoma. Semin Nucl Med 35:160-164, 2005. 10. Michelini S, Cardone M, Failla A, et al. Clinical staging. In Lee BB, Bergan J, Rockson SG, eds. Lymphedema: A Concise Compendium of Theory and Practice. London: Springer-Verlag, 2011. 11. Lawenda BD, Mondry TE, Johnstone PA. Lymphedema: a primer on the identification and management of a chronic condition in oncologic treatment. CA Cancer J Clin 59:8-24, 2009. 12. Rockson SG. Diagnosis and management of lymphatic vascular disease. J Am Coll Cardiol 52:799806, 2008.

K23346_Neligan_20_Causes_r3_0275-0290.indd 285

5/26/2015 9:22:00 AM

286

Part IV  Diagnosis of Lymphedema

13. Badger CM, Peacock JL, Mortimer PS. A randomized, controlled, parallel-group clinical trial comparing multilayer bandaging followed by hosiery versus hosiery alone in the treatment of patients with lymphedema of the limb. Cancer 88:2832-2837, 2000. 14. Camacho A, Jiménez F, De La Torre J, et al. Assembly of Bacillus subtilis phage phi29. 1. Mutants in the cistrons coding for the structural proteins. Eur J Biochem 73:39-55, 1977. 15. Ferrell RE, Kimak MA, Lawrence EC, et al. Candidate gene analysis in primary lymphedema. Lymphat Res Biol 6:69-76, 2008. 16. Mortimer PS, Rockson SG. New developments in clinical aspects of lymphatic disease. J Clin Invest 124:915-921, 2014. 17. Jacob AG, Driscoll DJ, Shaughnessy WJ, et al. Klippel-Trenaunay syndrome: spectrum and management. Mayo Clin Proc 73:28-36, 1998. 18. Denniston A. Turner’s syndrome. Lancet 358:2169-2170, 2001. 19. Connell F, Brice G, Mortimer P. Phenotypic characterization of primary lymphedema. Ann N Y Acad Sci 1131:140-146, 2008. 20. Evans AL, Brice G, Sotirova V, et al. Mapping of primary congenital lymphedema to the 5q35.3 region. Am J Hum Genet 64:547-555, 1999. 21. Brice G, Mansour S, Bell R, et al. Analysis of the phenotypic abnormalities in lymphoedema-​distichiasis syndrome in 74 patients with FOXC2 mutations or linkage to 16q24. J Med Genet 39:478-483, 2002. 22. Mellor RH, Brice G, Stanton AW, et al. Mutations in FOXC2 are strongly associated with primary valve failure in veins of the lower limb. Circulation 115:1912-1920, 2007. 23. Szuba A, Rockson SG. Lymphedema: classification, diagnosis and therapy. Vasc Med 3:145-156, 1998. 24. Smeltzer DM, Stickler GB, Schirger A. Primary lymphedema in children and adolescents: a follow-up study and review. Pediatrics 76:206-218, 1985. 25. Rockson SG, Rivera KK. Estimating the population burden of lymphedema. Ann N Y Acad Sci 1131:147-154, 2008. 26. Balzer K, Schonebeck I. [Edema after vascular surgery interventions and its therapy] Z Lymphol 17:4147, 1993. 27. Carrizo GJ, Livesay JJ, Luy L. Endoscopic harvesting of the greater saphenous vein for aortocoronary bypass grafting. Tex Heart Inst J 26:120-123, 1999. 28. Ibanez JP, Monteverde ML, Goldberg J, et al. Sirolimus in pediatric renal transplantation. Transplant Proc 37:682-684, 2005. 29. Aldrete JA, Couto da Silva JM. Leg edema from intrathecal opiate infusions. Eur J Pain 4:361-365, 2000. 30. Keiser PB, Nutman TB. Update on lymphatic filarial infections. Curr Infect Dis Rep 4:65-69, 2002. 31. Nutman TB. Lymphatic filariasis: new insights and prospects for control. Curr Opin Infect Diseases 14:539-546, 2001. 32. Global Programme to Eliminate Lymphatic Filariasis. Progress report on mass drug administration, 2010. Weekly Epidemiological Record/Health Section of the Secretariat of the League of Nations 86:377-388, 2011. 33. Dreyer G, Medeiros Z, Netto MJ, et al. Acute attacks in the extremities of persons living in an area endemic for bancroftian filariasis: differentiation of two syndromes. Trans R Soc Trop Med Hyg 93:413417, 1999. 34. Kumaraswami V. The clinical manifestations of lymphatic filariasis. In Nutman TB, ed. Lymphatic Filariasis Tropical Medicine: Science and Practice. London: Imperial College Press, 2000. 35. Shenoy RK, John A, Babu BS, et al. Two-year follow-up of the microfilaraemia of asymptomatic brugian filariasis, after treatment with two, annual, single doses of ivermectin, diethylcarbamazine and albendazole, in various combinations. Ann Trop Med Parasitol 94:607-614, 2000. 36. Dunyo SK, Nkrumah FK, Simonsen PE. A randomized double-blind placebo-controlled field trial of ivermectin and albendazole alone and in combination for the treatment of lymphatic filariasis in Ghana. Trans R Soc Trop Med Hyg 94:205-211, 2000.

K23346_Neligan_20_Causes_r3_0275-0290.indd 286

5/26/2015 9:22:00 AM

Chapter 20  Causes and Classification of Lymphatic Disorders

287

37. Hoerauf A, Volkmann L, Nissen-Paehle K, et al. Targeting of Wolbachia endobacteria in Litomosoides sigmodontis: comparison of tetracyclines with chloramphenicol, macrolides and ciprofloxacin. Trop Med Int Health 5:275-279, 2000. 38. Ryncarz RE, Heasley EC, Babinchak TJ. The clinical spectrum of nodular lymphangitis. Hosp Physician 35:63-66, 1999. 39. Wu JW, Chiles C. Lymphangitic carcinomatosis from prostate carcinoma. J Comput Assist Tomogr 23:761-763, 1999. 40. Kohmo S, Tachibana I, Osaki T, et al. Multiple organ mucosa-associated lymphoid tissue lymphoma presenting with lymphangitic pattern of spread in the lung. J Thoracic Oncol 2:1057-1059, 2007. 41. Murphy MJ, Kogan B, Carlson JA. Granulomatous lymphangitis of the scrotum and penis. Report of a case and review of the literature of genital swelling with sarcoidal granulomatous inflammation. J Cutan Pathol 28:419-424, 2001. 42. Van Kruiningen HJ, Colombel JF. The forgotten role of lymphangitis in Crohn’s disease. Gut 57:1-4, 2008. 43. Rosen T, Hwong H. Sclerosing lymphangitis of the penis. J Am Acad Dermatol 49:916-918, 2003. 44. Schirger A. Lymphedema. Cardiovasc Clin 13:293-305, 1983. 45. Kostman JR, DiNubile MJ. Nodular lymphangitis: a distinctive but often unrecognized syndrome. Ann Intern Med 118:883-888, 1993. 46. Lohrmann C, Foeldi E, Bartholomä JP, et al. Interstitial MR lymphangiography—a diagnostic imaging method for the evaluation of patients with clinically advanced stages of lymphedema. Acta Trop 104:8-15, 2007. 47. Garfein ES, Borud LJ, Warren AG, et al. Learning from a lymphedema clinic: an algorithm for the management of localized swelling. Plast Reconstr Surg 121:521-528, 2008. 48. Shin BW, Sim YJ, Jeong HJ, et al. Lipedema, a rare disease. Ann Rehabil Med 35:922-927, 2011. 49. Vignes S. [Lipedema: a misdiagnosed entity] J Mal Vasc 37:213-218, 2012. 50. Reich-Schupke S, Altmeyer P, Stucker M. Thick legs—not always lipedema. J Dtsch Dermatol Ges 11:225-233, 2013. 51. Szolnoky G, Varga E, Varga M, et al. Lymphedema treatment decreases pain intensity in lipedema. Lymphology 44:178-182, 2011. 52. Wagner S. Lymphedema and lipedema—an overview of conservative treatment. VASA 40:271-279, 2011. 53. Faul JL, Berry GJ, Colby TV, et al. Thoracic lymphangiomas, lymphangiectasis, lymphangiomatosis, and lymphatic dysplasia syndrome. Am J Respir Crit Care Med 161:1037-1046, 2000. 54. Ganesh C, Sangeetha GS, Narayanan V, et al. Lymphangioma circumscriptum in an adult: an unusual oral presentation. J Clin Imaging Sci 3:44, 2013. 55. Mordehai J, Kurzbart E, Shinhar D, et al. Lymphangioma circumscriptum. Pediatr Surg Int 13:208210, 1998. 56. Whimster IW. The pathology of lymphangioma circumscriptum. Br J Dermatol 94:473-486, 1976. 57. Chervenak FA, Isaacson G, Blakemore KJ, et al. Fetal cystic hygroma. Cause and natural history. N Engl J Med 309:822-825, 1983. 58. Radhakrishnan K, Rockson SG. The clinical spectrum of lymphatic disease. Ann N Y Acad Sci 1131:155184, 2008. 59. Marcucci G, Masi L, Carossino AM, et al. Cystic bone angiomatosis: a case report treated with aminobisphosphonates and review of the literature. Calcif Tissue Int 93:462-471, 2013. 60. Schajowicz F, Aiello CL, Francone MV, et al. Cystic angiomatosis (hamartous haemolymphagiomatosis) of bone. A clinicopathological study of three cases. J Bone Joint Surgery Br 60:100-106, 1978. 61. Boyle WJ. Cystic angiomatosis of bone. A report of three cases and review of the literature. J Bone Joint Surg Br 54:626-636, 1972. 62. Seckler SG, Rubin H, Rabinowitz JG. Systemic cystic angiomatosis. Am J Med 37:976-986, 1964. 63. Brunzell JD, Shankle SW, Bethune JE. Congenital generalized lipodystrophy accompanied by cystic angiomatosis. Ann Intern Med 69:501-516, 1968.

K23346_Neligan_20_Causes_r3_0275-0290.indd 287

5/26/2015 9:22:00 AM

288

Part IV  Diagnosis of Lymphedema

64. Albregts AE, Rapini RP. Malignancy in Maffucci’s syndrome. Dermatol Clin 13:73-78, 1995. 65. Kerr HD, Keep JC, Chiu S. Lymphangiosarcoma associated with lymphedema in a man with Maffucci’s syndrome. South Med J 84:1039-1041, 1991. 66. Jermann M, Eid K, Pfammatter T, et al. Maffucci’s syndrome. Circulation 104:1693, 2001. 67. Kinmonth JB. The Lymphatics: Surgery, Lymphography, and Diseases of the Chyle and Lymph Systems. London: Edward Arnold Publishers, 1982. 68. Manisali M, Ozaksoy D. Gorham disease: correlation of MR findings with histopathologic changes. Eur Radiol 8:1647-1650, 1998. 69. Patel DV. Gorham’s disease or massive osteolysis. Clin Med Res 3:65-74, 2005. 70. Mawk JR, Obukhov SK, Nichols WD, et al. Successful conservative management of Gorham disease of the skull base and cervical spine. Childs Nerv Syst 13:622-625, 1997. 71. Dunbar SF, Rosenberg A, Mankin H, et al. Gorham’s massive osteolysis: the role of radiation therapy and a review of the literature. Int J Radiat Oncol Biol Phys 26:491-497, 1993. 72. Heyd R, Micke O, Surholt C, et al. Radiation therapy for Gorham-Stout syndrome: results of a national patterns-of-care study and literature review. Int J Radiat Oncol Biol Phy 81:e179-e185, 2011. 73. Raboudi T, Bouchoucha S, Hamdi B, et al. Soft-tissue necrosis complicating tibial osteotomy in a child with Proteus syndrome. Orthop Traumatol Surg Res 100:247-250, 2014. 74. Biesecker L. The challenges of Proteus syndrome: diagnosis and management. Eur J Hum Genet 14:1151-1157, 2006. 75. Clark RD, Donnai D, Rogers J, et al. Proteus syndrome: an expanded phenotype. Am J Med Genet 27:99-117, 1987. 76. Kihiczak GG, Meine JG, Schwartz RA, et al. Klippel-Trenaunay syndrome: a multisystem disorder possibly resulting from a pathogenic gene for vascular and tissue overgrowth. Int J Dermatol 45:883890, 2006. 77. Mulliken JB, Glowacki J. Hemangiomas and vascular malformations in infants and children: a classification based on endothelial characteristics. Plast Reconstr Surg 69:412-422, 1982. 78. Huiras EE, Barnes CJ, Eichenfield LF, et al. Pulmonary thromboembolism associated with KlippelTrenaunay syndrome. Pediatrics 116:e596-e600, 2005. 79. Dubois J, Garel L, Grignon A, et al. Imaging of hemangiomas and vascular malformations in children. Acad Radiol 5:390-400, 1998. 80. Dubois J, Garel L. Imaging and therapeutic approach of hemangiomas and vascular malformations in the pediatric age group. Pediatr Radiol 29:879-893, 1999. 81. Kern S, Niemeyer C, Darge K, et al. Differentiation of vascular birthmarks by MR imaging. An investigation of hemangiomas, venous and lymphatic malformations. Acta Radiol 41:453-457, 2000. 82. Berry SA, Peterson C, Mize W, et al. Klippel-Trenaunay syndrome. Am J Med Genet 79:319-326, 1998. 83. Ring DS, Mallory SB. What syndrome is this? Klippel-Trenaunay syndrome. Pediatr Dermatol 9:8082, 1992. 84. Enjolras O, Mulliken JB. The current management of vascular birthmarks. Pediatr Dermatol 10:311313, 1993. 85. Rodrigues D, Bourroul ML, Ferrer AP, et al. Blue rubber bleb nevus syndrome. Rev Hosp Clin Fac Med Sao Paulo 55:29-34, 2000. 86. Ganesh R, Reddy M, Janakiraman L, et al. Blue rubber bleb nevus syndrome. Indian J Pediatr 81:317318, 2014. 87. Ertem D, Acar Y, Kotiloglu E, et al. Blue rubber bleb nevus syndrome. Pediatrics 107:418-420, 2001. 88. Chiu NT, Lee BF, Hwang SJ, et al. Protein-losing enteropathy: diagnosis with (99m)Tc-labeled human serum albumin scintigraphy. Radiology 219:86-90, 2001. 89. Hilliard RI, McKendry JB, Phillips MJ. Congenital abnormalities of the lymphatic system: a new clinical classification. Pediatrics 86:988-994, 1990. 90. Bhatnagar A, Kashyap R, Chauhan UP, et al. Diagnosing protein losing enteropathy. A new approach using Tc-99m human immunoglobulin. Clin Nucl Med 20:969-972, 1995.

K23346_Neligan_20_Causes_r3_0275-0290.indd 288

5/26/2015 9:22:01 AM

Chapter 20  Causes and Classification of Lymphatic Disorders

289

91. Strober W, Wochner RD, Carbone PP, et al. Intestinal lymphangiectasia: a protein-losing enteropathy with hypogammaglobulinemia, lymphocytopenia and impaired homograft rejection. J Clin Invest 46:1643-1656, 1967. 92. Donnelly JP, Rosenthal A, Castle VP, et al. Reversal of protein-losing enteropathy with heparin therapy in three patients with univentricular hearts and Fontan palliation. J Pediatr 130:474-478, 1997. 93. Warshaw AL, Waldmann TA, Laster L. Protein-losing enteropathy and malabsorption in regional enteritis: cure by limited ileal resection. Ann Surg 178:578-580, 1973. 94. Rychik J, Piccoli DA, Barber G. Usefulness of corticosteroid therapy for protein-losing enteropathy after the Fontan procedure. Am J Cardiol 68:819-821, 1991. 95. Rothman A, Snyder J. Protein-losing enteropathy following the Fontan operation: resolution with prednisone therapy. Am Heart J 121:618-619, 1991. 96. Fakhri A, Fishman EK, Jones B, et al. Primary intestinal lymphangiectasia: clinical and CT findings. J Comput Assist Tomogr 9:767-770, 1985. 97. Puri AS, Aggarwal R, Gupta RK, et al. Intestinal lymphangiectasia: evaluation by CT and scintigraphy. Gastrointest Radiol 17:119-121, 1992. 98. Laor T, Hoffer FA, Burrows PE, et al. MR lymphangiography in infants, children, and young adults. AJR Am J Roentgenol 171:1111-1117, 1998. 99. Yueh TC, Pui MH, Zeng SQ. Intestinal lymphangiectasia: value of Tc-99m dextran lymphoscintigraphy. Clin Nucl Med 22:695-696, 1997. 100. Bhatnagar A, Lahoti D, Singh AK, et al. Scintigraphic diagnosis of protein losing enteropathy using Tc-99m dextran. Clin Nucl Med 20:1070-1073, 1995. 101. Holt PR. Dietary treatment of protein loss in intestinal lymphangiectasia. The effect of eliminating dietary long chain triglycerides on albumin metabolism in this condition. Pediatrics 34:629-635, 1964. 102. Soto-Martinez M, Massie J. Chylothorax: diagnosis and management in children. Paediatr Respir Rev 10:199-207, 2009. 103. Johnson S. Rare diseases. 1. Lymphangioleiomyomatosis: clinical features, management and basic mechanisms. Thorax 54:254-264, 1999. 104. Travis WD, Usuki J, Horiba K, et al. Histopathological studies on lymphangioleiomyomatosis. In Moss J, ed. LAM and Other Diseases Characterized by Smooth Muscle Proliferation. Lung Biology in Health and Disease. New York: Marcel Dekker, 1999. 105. Johnson SR, Cordier JF, Lazor R, et al. European Respiratory Society guidelines for the diagnosis and management of lymphangioleiomyomatosis. Eur Respir J 35:14-26, 2010. 106. Derweduwen AM, Verbeken E, Stas M, et al. Extrapulmonary lymphangioleiomyomatosis: a wolf in sheep’s clothing. Thorax 68:111-113, 2013. 107. Chu SC, Horiba K, Usuki J, et al. Comprehensive evaluation of 35 patients with lymphangioleiomyomatosis. Chest 115:1041-1052, 1999. 108. Bosniak MA, Megibow AJ, Hulnick DH, et al. CT diagnosis of renal angiomyolipoma: the importance of detecting small amounts of fat. AJR Am J Roentgenol 151:497-501, 1988. 109. Bernstein SM, Newell JD Jr, Adamczyk D, et al. How common are renal angiomyolipomas in patients with pulmonary lymphangiomyomatosis? Am J Respir Crit Care Med 152:2138-2143, 1995. 110. Moss J, DeCastro R, Patronas NJ, et al. Meningiomas in lymphangioleiomyomatosis. JAMA 286:18791881, 2001. 111. Johnson SR, Clelland CA, Ronan J, et al. The TSC-2 product tuberin is expressed in lymphangioleiomyomatosis and angiomyolipoma. Histopathology 40:458-463, 2002. 112. Bissler JJ, Kingswood JC. Renal angiomyolipomata. Kid Int 66:924-934, 2004. 113. Ferrans VJ, Yu ZX, Nelson WK, et al. Lymphangioleiomyomatosis (LAM): a review of clinical and morphological features. J Nippon Med Sch 67:311-329, 2000.

K23346_Neligan_20_Causes_r3_0275-0290.indd 289

5/26/2015 9:22:01 AM

K23346_Neligan_20_Causes_r3_0275-0290.indd 290

5/26/2015 9:22:01 AM

C hapter 21 Basic Approaches to the Diagnosis of Lymphedema: Clinicians’ Perspective Yener Demirtas, Baris Yigit

K ey P oints • Lymphedema can be defined as the chronic accumulation of lymphatic fluid in the interstitial space between tissue cells.

Bas nici

• The differential diagnosis of edema requires a detailed medical history of the patient and a physical examination. Physician experience diagnosing lymphedema also plays a key role. • According to the 2013 Consensus Document of the International Society of Lymphology, lymphedema can be divided into four stages. • To establish an accurate diagnosis of lymphedema, the most important factors are the duration of the swelling, clinical findings, and whether the disease is unilateral or bilateral. Clinical tests, such as an MRI, CT, SPECT-CT, and isotopic lymphoscintigraphy, also play a crucial role.

Clinically, lymphedema can be described as chronic excessive fluid accumulation in the interstitial spaces between tissue cells. This deposition (which at least in the extremities is epifascial) swelling is a consequence of increased inflow or decreased outflow or both.1 The diagnosis of lymphedema is basically made by a familial and clinical history and physical examination. To do this well, the physician should have many prior assessments of clinically manifest lymphedema. The typical presentation of a patient with early to midstage lymphedema is with a combination of the following signs and symptoms: whole or partial limb swelling; tightness in the skin; pitting caused by rings, a watchband, tight socks, or underwear; and a history of recurrent infections or functional complaints, such as heaviness, fatigue, and difficulty moving joints.2,3 Most patients will also have a history of injury or damage (for example, from cancer treatment) to the lymphatic

291

K23346_Nelgian_21_Basic Approach_r3_dc_0291-0298.indd 291

5/26/2015 9:37:04 AM

292

Part IV  Diagnosis of Lymphedema

system, because secondary lymphedema is the most common form of lymphedema, at least in Western populations. However, we are now aware that for some of these patients, there may be an underlying primary component. Furthermore, obesity, infections, and a history of radiotherapy significantly increase the risk of lymphedema. Patients with primary lymphedema may have a family history; however, sporadic forms of primary lymphedema are more common. In these situations, the diagnosis of primary lymphedema is one of exclusion.3

Differential Diagnosis There are many reasons for a swollen limb, and an accurate and appropriate diagnosis is critical to ensure the appropriate treatment. The differential diagnosis of edema requires a detailed medical history of the patient, physical examination, and occasionally special laboratory tests, such as an MRI and ultrasonography. Although distinguishing, the duration and distribution of the edema and the presence of dermatologic changes depend on the reason for the edema.4,5 According to the 2013 Consensus Document of the International Society of Lymphology,6 lymphedema has four stages. Stage 0 refers to the latent or subclinical period. Stage I refers to the early phase of swelling. Stage II swelling is accurate, and limb elevation does not reduce the swelling alone, although it does in stage I. Stage III refers to lymphostatic elephantiasis. If the duration of the edema is less than 2 weeks and also unilateral with the presentation of pain, it may be related to an acute condition, such as deep vein thrombosis, cellulitis, or a ruptured Baker cyst.7 If the edema is gradual and progressive over a few months (suggesting chronicity) and is unilateral without pain, then a possible diagnosis may be chronic venous insufficiency, external venous compression caused by a tumor of the iliac region of the lower extremity, or tumors of the soft tissue or vascular structures.8 However, lymphedema (primary or secondary) cannot be easily excluded. If the beginning of the edema is gradual, progressive, and bilateral, possible causes include congestive heart failure, glomerulonephritis, cirrhosis, lipedema, chronic venous insufficiency, or a malignancy in the pelvis, abdomen, or retroperitoneal space.7,8 Of course, it could also be bilateral lymphedema. Table 21-1 defines the differential diagnosis. Rarely, patients with long-standing lymphedema will present with lymphangiosarcoma, an aggressive tumor with a 5-year survival rate of less than 10%. This complication was first reported by Stewart and Treves in 1948 in patients with postmastectomy lymphedema and is referred to as Stewart-Treves syndrome. In this case, patients present with red or purple nodules in the diseased tissues and are most commonly treated with amputation (Fig. 21-1).

K23346_Nelgian_21_Basic Approach_r3_dc_0291-0298.indd 292

5/26/2015 9:37:04 AM

Chapter 21  Basic Approaches to the Diagnosis of Lymphedema: Clinicians’ Perspective

293

TABLE 21-1  Differential Diagnosis of Lymphedema Situation

History

Clinical Findings

Unilateral/Bilateral

Possible Diagnosis

Acute

Short history (less than 2 wk)

Pain

Unilateral

Deep vein thrombosis, cellulitis, or ruptured Baker cyst

Chronic

Gradual or progressive

No pain

Unilateral

Chronic venous insufficiency, lymphedema (primary or secondary), external venous compression, or soft tissue or vascular tumor

Chronic

Gradual and progressive

No pain

Bilateral

Congestive heart failure, glomerulonephritis, cirrhosis, lipedema, bilateral chronic venous insufficiency, bilateral lymphedema, or malignancy (pelvic, abdominal, or retroperitoneal)

FIG. 21-1  A patient with postmastectomy lymphedema with Stewart-Treves syndrome.

Diagnosis of Lymphedema Although it is not difficult to establish a diagnosis of lymphedema with clinical findings and questioning the medical history of patients with advanced lymphedema,9 it can be difficult to distinguish other reasons for the edema, especially in the early stages, when mild and/or intermittent swelling is present. Some physical findings make the discrimination easy, such as the peau d’orange appearance of the skin (cutaneous and subcutaneous fibrosis). The Stemmer sign (an inability to pick up a skin fold of the dorsum of the second toe between the thumb and forefinger) can help distinguish lymphedema from other causes of the swelling, particularly in the middle or later stages, although it can be positive in the normal population.10,11

K23346_Nelgian_21_Basic Approach_r3_dc_0291-0298.indd 293

5/26/2015 9:37:04 AM

294

Part IV  Diagnosis of Lymphedema

15 minutes

60 minutes

120 minutes

FIG. 21-2  Typical lymphoscintigraphic findings of a unilateral lower extremity lymphedema. The images were scanned 15, 60, and 120 minutes after injection of the tracer. Lymph drainage without pathologic findings and clearly visible inguinal lymph nodes in the left (normal) leg is seen. The right leg shows diffuse distribution of the radiopharmaceutical in the lower leg (dermal backflow). The lymphatic pathways are not visualized, and the inguinal nodes are only noticeable at 120 minutes.

Additional tests may be required to confirm the diagnosis of lymphedema, because the physical examination and medical history do not always provide the whole, accurate picture. The diagnosis of lymphedema can be strengthened by tests such as isotopic lymphoscintigraphy, indirect and direct lymphography, lymphatic capillaroscopy, MRI, axial tomography, ultrasonography, and more recently the use of indocyanine green (ICG). The mechanism of isotopic lymphoscintigraphy is monitoring the radio-labeled macromolecule with a gamma-camera after the intradermal injection of ICG contrast dye into the interdigital area of the toes or fingers. In a healthy lymphatic system, major lymphatic trunks and nodes will be rapidly seen. A common finding in lymphedema is an inability to visualize the trunks or delayed monitoring of tracer at the lymphatic trunks and lymph nodes. The presence of dermal backflow, which occurs from the escape of the radio-labeled molecule from the major lymphatic trunks to the skin, is another common finding in lymphedema12-15 (Fig. 21-2). Dermal backflow is frequently seen in secondary lymphedema, which is indicative of a proximal obstruction. Lymphangiography is another test that is generally not used much today because of technical difficulties, such as the detection and cannulation of the lymphatics. Another reason is that the contrast dye can injure the remaining lymphatic vessels and thus worsen the lymphedema. One

K23346_Nelgian_21_Basic Approach_r3_dc_0291-0298.indd 294

5/26/2015 9:37:05 AM

Chapter 21  Basic Approaches to the Diagnosis of Lymphedema: Clinicians’ Perspective

295

of the new techniques, ICG fluorescence, is very valuable for diagnosis and is also used for lymphatic venous anastomosis. With this test the lymphatic pathways, obliteration level, and dermal backflow can easily be detected. The diagnosis and structural tissue changes of lymphedema and their quantification can also be performed with various noninvasive methods, including perometry,16,17 tissue tonometry,16 bioimpedance spectroscopy,18-20 tissue dielectric constants,20 and other radiologic imaging techniques. Perometry is a method for calculating limb volumes and relies on the use of infrared scanning technology to estimate limb cross-sectional diameters at multiple intervals. Bioimpedance measures the rate of electrical current transmission through tissues and can estimate fluid content in a lymphedematous limb when compared with the normal limb. This technique is particularly helpful in early stage lymphedema, because it can detect fluids long before they are often clinically discernible. To note and document the structural changes, MR and CT imaging and single photon emission computed tomography (SPECT)–CT imaging are the best evaluation tests.21 Radiographic discrimination of lymphedema involves the absence of the edema within the muscular compartment. Fluid collections within the subfascial compartment may also be seen, even if the swelling is presumably limited by the inelastic deep fascia investing the muscle.22 Another radiographic finding is the honeycomb distribution of the edema, which is characteristic of lymphedema (not seen in other edematous situations) within the epifascial plane, along with thickening of the skin. SPECT-CT images are obtained with overlapped images of the lymphoscintigraphic SPECT and anatomic CT data. Tissue tonometry and indurometry, bioimpedance analysis, and tissue dielectric constants are easy, noninvasive tests that are also useful to detect the response to treatment.16-20 Finally, ultrasound can be used to evaluate lymphedema by correlating the thickness of the subcutaneous tissue with the progression of lymphedema and fibrosis.3

Conclusion The diagnosis of a typical patient with lymphedema is often quite easy for a physician who is experienced in recognizing lymphedema when a thorough medical history is obtained and a physical examination performed. However, because of the lack of general awareness of the disease among surgeons, most patients, especially those with lymphedema of the lower extremity, may still visit many physicians before they are finally diagnosed with lymphedema. Clinically, lymphedema can be described as the chronic accumulation of lymphatic fluid in the interstitial space between tissue cells. For a diagnosis, an evaluation of the patient history and a physical examination are almost sufficient. Lymphedema has four stages, 0 through III, based on the consensus document of the International Society of Lymphology. A differential diagnosis should be performed carefully. The most important factors to establish an accurate diagnosis of lymphedema are the duration of the swelling, clinical findings, and whether the disease is unilateral or bilateral. Clinical tests such as an MRI, CT, SPECT-CT, and isotopic lymphoscintigraphy are also important to establish a diagnosis.

K23346_Nelgian_21_Basic Approach_r3_dc_0291-0298.indd 295

5/26/2015 9:37:05 AM

296

Part IV  Diagnosis of Lymphedema

C linical P earls • Twenty percent of patients with lymphedema referred to our clinic by other physicians have lipedema, not lymphedema. • During an examination of a patient with unilateral lymphedema of the lower extremity, the surgeon should remember that 25% of these unilateral cases are or will be bilateral. We have diagnosed many patients with bilateral disease who were unaware of the earlier stage lymphedema at the contralateral lower extremity. • The surgeon should always exclude a recurrence in patients with an oncologic history. • The severity or stage of the disease is not always related to the duration of the lymphedema.

R EFERENCES 1. Daroczy J. Pathology of chronic lymphedema. Lymphology 6:91-106, 1994. 2. Warren AG, Brorson H, Borud LJ, et al. Lymphedema: a comprehensive review. Ann Plast Surg 59:464472, 2007. 3. Levine ST, Chang DW, Mehrara BJ. Lymphedema: diagnosis and treatment. In Thorne CHM, ed. Grabb and Smith’s Plastic Surgery, ed 7. Philadelphia: Lippincott William & Wilkins, 2014. 4. Dreyer G, Addiss D, Dreyer P, et al. Basic Lymphoedema Management, Treatment and Prevention of Problems Associated with Lymphatic Filariasis. Hollis, NH: Hollis Publishing, 2002. 5. Herpertz U. Lymphödem und Erysipel. Fortschr Med 116:36-40, 1998. 6. International Society of Lymphology. The diagnosis and treatment of peripheral lymphedema: 2013 Consensus Document of the International Society of Lymphology. Lymphology 46:1-11, 2013. 7. Browse N, Burnand K, Mortimer P, eds. Diseases of the Lymphatics. London: Arnold, 2003. 8. Kim DI, Huh S, Lee SJ, et al. Excision of subcutaneous tissue and deep muscle fascia for advanced lymphedema. Lymphology 31:190-194, 1998. 9. Rockson SG, Miller LT, Senie R, et al. American Cancer Society lymphedema workshop. Workgroup III: diagnosis and management of lymphedema. Cancer 83(Suppl 12):2882-2885, 1998. 10. Stemmer R. [A clinical symptom for the early and differential diagnosis of lymphedema] Vasa 5:261262, 1976. 11. Pannier F, Hoffmann B, Stang A, et al. Prevalence of Stemmer’s sign in the general population. Results from the Bonn Vein Study. Phlebologie 6:289-292, 2007. 12. Cambria RA, Gloviczki P, Naessens JM, et al. Noninvasive evaluation of the lymphatic system with lymphoscintigraphy: a prospective, semiquantitative analysis in 386 extremities. J Vasc Surg 18:773782, 1993. 13. Ter SE, Alavi A, Kim CK, et al. Lymphoscintigraphy. A reliable test for the diagnosis of lymphedema. Clin Nucl Med 18:646-654, 1993. 14. Case TC, Witte CL, Witte MH, et al. Magnetic resonance imaging in human lymphedema: comparison with lymphangioscintigraphy. Magn Reson Imaging 10:549-558, 1992. 15. Stanton AW, Mellor RH, Cook GJ, et al. Impairment of lymph drainage in subfascial compartment of forearm in breast cancer-related lymphedema. Lymphat Res Biol 1:121-132, 2003. 16. Piller NB, Clodius L. The use of a tissue tonometer as a diagnostic aid in extremity lymphoedema: a determination of its conservative treatment with benzo-pyrones. Lymphology 9:127-132, 1976.

K23346_Nelgian_21_Basic Approach_r3_dc_0291-0298.indd 296

5/26/2015 9:37:05 AM

Chapter 21  Basic Approaches to the Diagnosis of Lymphedema: Clinicians’ Perspective

297

17. Mikes DM, Cha BA, Dym CL, et al. Bioelectrical impedance analysis revisited. Lymphology 32:157-165, 1999. 18. Bunce IH, Mirolo BR, Hennessy JM, et al. Post-mastectomy lymphoedema treatment and measurement. Med J Aust 161:125-128, 1994. 19. Ward LC, Bunce IH, Cornish BH, et al. Multi-frequency bioelectrical impedance augments the diagnosis and management of lymphoedema in post-mastectomy patients. Eur J Clin Invest 22:751-754, 1992. 20. Mayrovitz HN, Davey S, Shapiro E. Localized tissue water changes accompanying one manual lymphatic drainage (MLD) therapy session assessed by changes in tissue dielectric constant in patients with lower extremity lymphedema. Lymphology 41:186-188, 2008. 21. Vaughan BF. CT of swollen legs. Clin Radiol 41:24-30, 1990. 22. Clodius L, Deak L, Piller NB. A new instrument for the evaluation of tissue tonicity in lymphoedema. Lymphology 9:1-5, 1976.

K23346_Nelgian_21_Basic Approach_r3_dc_0291-0298.indd 297

5/26/2015 9:37:05 AM

K23346_Nelgian_21_Basic Approach_r3_dc_0291-0298.indd 298

5/26/2015 9:37:05 AM

C hapter 22 Biomarkers Kerstin Buttler, Jörg Wilting

K ey P oints • Although the ideal biomarker showing the complete lymphatic vascular system has not yet been discovered, a combination of markers can be used to reliably detect the lymphatics. • In comparison with the blood vascular tree, there is heterogeneity of lymphatic endothelial cells in the lymphatic vascular network.

Bio

• The most constant marker to detect lymphatic vessels in human tissues appears to be double staining with antibodies against CD31 and PROX1.

It is highly likely that the lymphatic vascular system was already known to Hippocrates of Kos (about 460 to 377 BC). The Alexandrian school (about 300 BC to 600 AD) called the lymphatic vessels ductus lactei.1 Gasparo Asellius2 began the “reinvestigation” of the lymphatics in dogs, and Jean Pecquet3 and Olof Rudbeck4 followed soon thereafter in humans. Three hundred years later, the invention of the electron microscope clarified the cellular nature of the delicate wall of the initial lymphatics.5 However, efficient biomedical and cellular studies on the development and function of the lymphatics were hampered by the lack of reliable biomarkers of lymphatic endothelial cells (LECs) in health and disease. The first molecular LEC markers were found more or less by chance. However, they were the prerequisite to distinguish between LECs and blood vascular endothelial cells (BECs), and they enabled the purification of LECs in vitro and their subsequent global characterization at RNA and protein levels. Recent studies show the heterogeneity of LECs in the lymphatic vascular tree (initial lymphatics, collectors, lymph nodes, and trunks) and the organ-specific behavior of lymphatics. Therefore the ideal marker showing the complete lymphatic vascular system and differentiating it from blood vessels has not yet been found. Nevertheless, a combination of markers can be used to reliably detect the lymphatics. In this chapter we describe the molecules that have been used most widely to characterize LECs.

299

K23346_Neligan_22_Biomarkers_r3_dc_0299-0308.indd 299

5/26/2015 9:38:44 AM

300

Part IV  Diagnosis of Lymphedema

Ecto-59-Nucleotidase Enzyme histochemical staining for ecto-59-nucleotidase (NT5E, cluster of differentiation 73 [CD73]) was one of the first markers to identify the lymphatics, and staining for alkaline phosphatase activity was used in parallel to identify blood vessels.6 Ecto-59-nucleotidase is a glycosylphosphatidylinositol-anchored surface protein, which, in addition to other functions, catalyzes the hydrolysis of purine 59-nucleotide to nucleoside and orthophosphate in water. It is part of the cascade that completely hydrolyzes extracellular adenosine triphosphate to adenosine. It has anticoagulant activity and can be found in snake venom.7 However, NT5E and alkaline phosphatase are not specifically expressed in LECs or BECs, and the method relies on quantitative rather than qualitative differences. Immunostaining for NT5E confirms expression in both types of vessels, with more prominent signals in the lymphatics (Fig. 22-1). Nevertheless, in combination with other techniques, NT5E can be a useful marker of the lymphatics.

CD31 and PROX1 Double Staining Double immunostaining with antibodies against cluster of differentiation 31 (CD31) (platelet endothelial cell adhesion molecule 1 [PECAM1]) and PROX1 is the most reliable tool to detect lymphatic vessels in human tissues (Fig. 22-2). This method detects lymphatics in human fetuses and the tissues of healthy adults, including aged adults and patients with lymphedema. It also differentiates lymphangiomas from hemangiomas,8 although each single marker is not specifically expressed in LECs only. PECAM1 is an adhesion molecule of the immunoglobulin superfamily. It is located in the cell membrane of platelets, monocytes, neutrophils and the subsets of T cells.9 PECAM1 is highly enriched on the surface of BECs and to a lesser extent is expressed on LECs. It is a regulator of leukocyte diapedesis, but its functions are not completely understood, because it may have both proinflammatory and antiinflammatory functions.10 The homeobox-containing transcription factor Prox1 is the mammalian homolog of the Drosophila gene prospero. Prox1 is essential for the development of the lymphatic vascular system. In Prox1 knockout mice, the lymphatics do not develop, whereas the blood vessels are obviously normal.11 Because of multiple defects, the mice die during the early stages of development. In LECs, PROX1 is located in the nucleus and thereby facilitates the counting of LEC numbers in histologic sections. Costaining with CD31 must be performed, because PROX1 is not endothelial cell specific but is expressed in other cell types, including hepatocytes, pancreatic epithelium, cardiomyocytes, lens, retina, spinal, and vegetative ganglia.12,13 PROX1 is expressed in LECs but not in BECs. The only exception from this rule seems to be the valve-forming endothelial cells in the blood vascular system: the concave side of the cardiac valves and venous valves.12,14 Nevertheless, the combination of CD31 and PROX1 is an excellent marker for LECs in vivo both in the initial lymphatics and lymphatic collectors (see Fig. 22-2) and in vitro.

K23346_Neligan_22_Biomarkers_r3_dc_0299-0308.indd 300

5/26/2015 9:38:44 AM

301

Chapter 22  Biomarkers

L

L

100 mm

FIG. 22-1  Immunohistochemical staining for ecto-59-nucleotidase (NT5E) of the human colon. Note the strong expression in lymphatic vessels (L) and weak expression in other tissues.

A

L L L

L

L L L

L 100 mm

B

C

LC

LCy

50 mm

50 mm

FIG. 22-2  Immunofluorescence staining of human tissues with antibodies against CD31 (green) and PROX1 (red). Counterstaining of nuclei with DAPI (blue). A, Foreskin of a 2-year-old boy. Numerous initial lymphatics (L) are weakly positive for CD31 and show nuclear PROX1 expression. Blood vessels are CD31 positive (strong) and PROX1 negative. B, Lymphatic collector (LC) of an adult. LECs are positive for CD31 and PROX1. C, Lymphatic cyst (LCy) of an infant with multiple lymphangiomas. LECs are positive for CD31 and PROX1.

K23346_Neligan_22_Biomarkers_r3_dc_0299-0308.indd 301

5/26/2015 9:38:45 AM

302

Part IV  Diagnosis of Lymphedema

Hyaluronan Receptor LYVE1 LYVE1 is a transmembrane glycoprotein and a homolog of the hyaluronan receptor CD44.15 It is highly expressed in the initial lymphatics of the skin and other organs (Fig. 22-3) but inconsistently expressed in lymphatic collectors and lymph node sinusoids.16,17 In our experience LYVE1 is an excellent marker of the lymphatics in the mouse, but in the human we have observed weak staining also in veins. Other BECs that express LYVE1 are the sinusoids of the liver and spleen, high endothelial venules, and the syncytiotrophoblast of the placenta.18,19 A large number of scattered mesenchymal cells, many of which are obviously macrophages, are positive for LYVE1.20 It has been suggested that the physiologic function of LYVE1 resides in the turnover of hyaluronan. Hyaluronan is an important component of the extracellular matrix and is abundantly expressed in the dermis. Its main degradation sites are the lymph nodes and liver, and its transport to these organs is obviously by the lymphatic vessels.18 However, no specific alterations of the hyaluronan content were found in Lyve1 knockout mice.21

Podoplanin Podoplanin (also known as OTS-8, T1-alpha, E11 antigen) is a 43-kDa surface glycoprotein. The name refers to its expression in kidney podocytes, but its highly restricted expression in LECs and not in BECs was soon recognized.22 In addition to LECs, podoplanin is found in basal cells of the epidermis, mesothelium, type I alveolar cells, choroid plexus, cholangiocytes, and osteoblasts. In some cases it is expressed in plasma cells, Schwann cells, and fibrocytes. Podoplanin is useful in the diagnosis of lymphangiomas but should be used in combination with other markers.16,18,23 In the initial lymphatics, podoplanin is coexpressed with LYVE1 (Fig. 22-4). The staining with anti-podoplanin antibodies is highly similar to that of the antibody D2-40 raised against the 40-kDa oncofetal antigen M2A.24 D2-40 marks the initial lymphatics intensely (Fig. 22-5), but not the lymphatic collectors in all cases and lymph node sinuses only weakly. Blood vessels, including those of the liver and spleen, are not detected by the antibody. However, D2-40

FIG. 22-3  Immunohistochemical staining of human foreskin with anti-LYVE1 antibodies. LYVE1 (black signal) marks the initial lymphatics.

60 mm

K23346_Neligan_22_Biomarkers_r3_dc_0299-0308.indd 302

5/26/2015 9:38:45 AM

303

Chapter 22  Biomarkers

A

B

DAPI

C

Podoplanin

D

70 mm LYVE1

Merged

FIG. 22-4  Immunofluorescence staining of human foreskin with antibodies against podoplanin (green) and LYVE1 (red). A, Counterstaining of nuclei with DAPI (blue). B, Anti-podoplanin. C, Anti-LYVE1. D, Merged picture. Note the double labeling of the initial lymphatics.

50 mm

FIG. 22-5  Immunofluorescence staining of human foreskin with D2-40 antibodies (magenta). The initial lymphatics are positive. In addition, we observed staining of the perineural sheath of the peripheral nerves (not shown).

K23346_Neligan_22_Biomarkers_r3_dc_0299-0308.indd 303

5/26/2015 9:38:47 AM

304

Part IV  Diagnosis of Lymphedema

25 mm

FIG. 22-6  Immunofluorescence staining of lymphangioma tissue from an infant with antibodies against VEGFR-3 (green) and PROX1 (magenta). Lymphatic vessels are positive for the two markers.

also recognizes the mesothelium, follicular dendritic cells, and fibroblasts of scar tissue, basal cells of the epidermis and prostate, myoepithelial cells of the mammary gland, ependymal cells, and choroid plexus epithelium.16 In regard to neoplastic tissues, D2-40 marks gastrointestinal stroma tumors, seminomas, numerous brain tumors, and mesotheliomas.16

Vascular Endothelial Growth Factor Receptor 3 Vascular endothelial growth factor receptor 3 (VEGFR-3) is a receptor of the vascular endothelial growth factor (VEGF) family of endothelial growth factors. It is typically expressed on LECs and binds the lymphangiogenic growth factors VEGF-C and VEGF-D with high affinity.25 In addition to the transmembrane form of the receptor, an endogenous-secreted splice variant of VEGFR-3 was recently found, which acts as an inhibitor of lymphangiogenesis.26 VEGFR-3 is very important in the development of the lymphatics, and heterozygous mutations affecting the tyrosine kinase domain of the VEGFR-3 (FLT4) gene are the leading causes of the development of primary lymphedema.27 In early embryos, before the development of the lymphatics, VEGFR-3 is expressed in blood vessels. Its great importance in the development of early blood vessels is emphasized by cardiovascular failure and the early death of Flt4 knockout mice.28 Later in development, VEGFR-3 becomes restricted to the lymphatics and high endothelial venules. However, in pathologic tissues (solid tumors and hemangiomas), it becomes upregulated in subsets of blood vessels, reflecting the early embryonic status.29,30 Additional markers are then needed to unequivocally identify the lymphatics (Fig. 22-6).

Mannose Receptor, C Type 1 High expression of mannose receptor, C type 1 (MRC1) (also known as MRC1L1, CD206, CLEC13D) was found by gene microarray analyses in human LECs,31-33 although the detection of MRC1 at the protein level may be difficult in LECs. MRC1 is an endocytotic receptor for highmannose structures on the surface of pathogens (viruses, bacteria, and fungi). After non-selfrecognition, they can be neutralized by macrophages via phagocytic engulfment. Endogenous

K23346_Neligan_22_Biomarkers_r3_dc_0299-0308.indd 304

5/26/2015 9:38:47 AM

Chapter 22  Biomarkers

305

self-ligands include L-selectin, which is implicated in cell migration.34 The fact that Mrc1 knock­ out mice do not seem to have higher susceptibility for infection suggests that MRC1 functions can be compensated by other surface receptors.35 The highest levels of MRC1 are expressed during the resolution of inflammation.36 In addition, MRC1 has been implicated in antigen presentation.37 The expression of MRC1 on LECs, macrophages, and dendritic cells has led to the development of the clinical lymphatic tracer Lymphoseek, a radiolabeled (technetium-99m) macromolecule of multiple mannose units, which may be useful in the search for draining (sentinel) lymph nodes in cancer patients.38

Conclusion Studies on the lymphatic vascular system were characterized by great discontinuity during past centuries. After seminal documented studies on macroscopic aspects of the lymphatics in the seventeenth century,2,4 intense disputations on their development in the late nineteenth and early twentieth centuries,39,40 and clarification of their cellular composition in the 1960s,5 the era of molecular lymphology began in the 1990s with the detection of lymphangiogenic growth factors and their receptors.41-44 The identification of biomarkers of LECs facilitated their isolation and global characterization at RNA and protein levels and pointed to their distinction from BECs. Compared with the blood vascular tree, there is heterogeneity of LECs in the lymphatic vascular network. LECs of the initial lymphatics, collectors, and lymph node sinuses are molecularly distinct, and within the lymph nodes, even the parietal and visceral linings of the marginal sinuses are heterogeneous. The most constant marker for LECs seems to be double staining with the antibodies against CD31 and PROX1. We also learned that there is organ specificity of lymphatics; the visceral lymphatics show higher expression of the VEGF-C coreceptor neuropilin-2 compared with the dermal lymphatics,45 whereas the latter seems more dependent on signaling via Wnt5a.46 It is hoped that lymphedema and cancer patients will benefit from the translation of the data into the clinical realm.

R EFERENCES 1. Rusznyák I, Földi M, Szabó G. Lymphologie: Physiologie und Pathologie der Lymphgefässe und des Lymphkreislaufes. Stuttgart: Gustav Fischer, 1969. 2. Asellius G. De Lacteibus Sive Lacteis Venis Quarto Vasorum Mesaroicum Genere Novo Invente Gasp. Asellii Cremonesis Antomici Ticiensis Qua Sententiae Anatomicae multae, nel Perperam Receptae Illustrantur. Mediolani, apud Jo Baptistam Bidellium, 1627. 3. Pecquet J. Experimenta Nova Anatomica, Quibus Incognitum Chyli Receptaculum, et ab eo per Thoracem in Ramos Usque Subclavios Vasa Lactea Deteguntur. Montpellier, France, 1651. 4. Rudbeck O. Nova Exercitatio Anatomica Exhibens Ductus Hepaticos Aquosos et Vasa Glandularum Serosa, Nunc Primum Inventa, Aeneisque Figuris Delineata. Arosiae, Sweden, 1653. 5. Casley-Smith JR, Florey HW. The structure of normal small lymphatics. Q J Exp Physiol Cogn Med Sci 46:101-106, 1961. 6. Kato S, Miyauchi R. Enzyme-histochemical visualization of lymphatic capillaries in the mouse tongue: light and electron microscopic study. Okajimas Folia Anat Jpn 65:391-403, 1989. 7. Dhananjaya BL, Nataraju A, Rajesh R, et al. Anticoagulant effect of Naja naja venom 59nucleotidase: demonstration through the use of novel specific inhibitor, vanillic acid. Toxicon 48:411-421, 2006.

K23346_Neligan_22_Biomarkers_r3_dc_0299-0308.indd 305

5/26/2015 9:38:47 AM

306

Part IV  Diagnosis of Lymphedema

8. Wilting J, Papoutsi M, Christ B, et al. The transcription factor Prox1 is a marker for lymphatic endothelial cells in normal and diseased human tissues. FASEB J 16:1271-1273, 2002. 9. Newman PJ, Berndt MC, Gorski J, et al. PECAM-1 (CD31) cloning and relation to adhesion molecules of the immunoglobulin gene superfamily. Science 247:1219-1222, 1990. 10. Privratsky JR, Newman DK, Newman PJ. PECAM-1: conflicts of interest in inflammation. Life Sci 87:69-82, 2010. 11. Wigle JT, Oliver G. Prox1 function is required for the development of the murine lymphatic system. Cell 98:769-778, 1999. 12. Rodriguez-Niedenführ M, Papoutsi M, Christ B, et al. Prox1 is a marker of ectodermal placodes, endodermal compartments, lymphatic endothelium and lymphangioblasts. Anat Embryol (Berl) 204:399406, 2001. 13. Tomarev SI, Zinovieva RD, Chang B, et al. Characterization of the mouse Prox1 gene. Biochem Biophys Res Commun 248:684-689, 1998. 14. Bazigou E, Makinen T. Flow control in our vessels: vascular valves make sure there is no way back. Cell Mol Life Sci 70:1055-1066, 2013. 15. Banerji S, Ni J, Wang SX, et al. LYVE-1, a new homologue of the CD44 glycoprotein, is a lymph-specific receptor for hyaluronan. J Cell Biol 144:789-801, 1999. 16. Kaiserling E. Morphologische und funktionelle Aspekte des normalen und pathologisch Veränderten lymphatischen Gewebes. In Földi M, Földi E, eds. Lehrbuch der Lymphologie. Munich: Elsevier, 2010. 17. Kasten P, Schnöink G, Bergmann A, et al. Similarities and differences of human and experimental mouse lymphangiomas. Dev Dyn 236:2952-2961, 2007. 18. Sleeman JP, Krishnan J, Kirkin V, et al. Markers for the lymphatic endothelium: in search of the holy grail? Microsc Res Tech 55:61-69, 2001. 19. Wrobel T, Dziegiel P, Mazur G, et al. LYVE-1 expression on high endothelial venules (HEVs) of lymph nodes. Lymphology 38:107-110, 2005. 20. Chen L, Cursiefen C, Barabino S, et al. Novel expression and characterization of lymphatic vessel endothelial hyaluronate receptor 1 (LYVE-1) by conjunctival cells. Invest Ophthalmol Vis Sci 46:45364540, 2005. 21. Gale NW, Prevo R, Espinosa J, et al. Normal lymphatic development and function in mice deficient for the lymphatic hyaluronan receptor LYVE-1. Mol Cell Biol 27:595-604, 2007. 22. Breiteneder-Geleff S, Soleiman A, Kowalski H, et al. Angiosarcomas express mixed endothelial phenotypes of blood and lymphatic capillaries: podoplanin as a specific marker for lymphatic endothelium. Am J Pathol 154:385-394, 1999. 23. Schacht V, Ramirez MI, Hong YK, et al. T1alpha/podoplanin deficiency disrupts normal lymphatic vasculature formation and causes lymphedema. EMBO J 22:3546-3556, 2003. 24. Marks A, Sutherland DR, Bailey D, et al. Characterization and distribution of an oncofetal antigen (M2A antigen) expressed on testicular germ cell tumours. Br J Cancer 80:569-578, 1999. 25. Tammela T, Alitalo K. Lymphangiogenesis: Molecular mechanisms and future promise. Cell 140:460476, 2010. 26. Singh N, Tiem M, Watkins R, et al. Soluble vascular endothelial growth factor receptor 3 is essential for corneal alymphaticity. Blood 121:4242-4249, 2013. 27. Evans AL, Bell R, Brice G, et al. Identification of eight novel VEGFR-3 mutations in families with primary congenital lymphoedema. J Med Genet 40:697-703, 2003. 28. Dumont DJ, Jussila L, Taipale J, et al. Cardiovascular failure in mouse embryos deficient in VEGF receptor-3. Science 282:946-949, 1998. 29. Partanen TA, Alitalo K, Miettinen M. Lack of lymphatic vascular specificity of vascular endothelial growth factor receptor 3 in 185 vascular tumors. Cancer 86:2406-2412, 1999. 30. Wilting J, Papoutsi M, Othman-Hassan K, et al. Development of the avian lymphatic system. Microsc Res Tech 55:81-91, 2001.

K23346_Neligan_22_Biomarkers_r3_dc_0299-0308.indd 306

5/26/2015 9:38:47 AM

Chapter 22  Biomarkers

307

31. Hirakawa S, Hong YK, Harvey N, et al. Identification of vascular lineage-specific genes by transcriptional profiling of isolated blood vascular and lymphatic endothelial cells. Am J Pathol 162:575-586, 2003. 32. Petrova TV, Mäkinen T, Mäkelä TP, et al. Lymphatic endothelial reprogramming of vascular endothelial cells by the Prox-1 homeobox transcription factor. EMBO J 21:4593-4599, 2002. 33. Wilting J, Becker J, Buttler K, et al. Lymphatics and inflammation. Curr Med Chem 16:4581-4592, 2009. 34. Gordon S. Pattern recognition receptors: doubling up for the innate immune response. Cell 111:927930, 2002. 35. Gazi U, Martinez-Pomares L. Influence of the mannose receptor in host immune responses. Immunobiology 214:554-561, 2009. 36. Lee SJ, Evers S, Roeder D, et al. Mannose receptor-mediated regulation of serum glycoprotein homeostasis. Science 295:1898-1901, 2002. 37. Stahl PD, Ezekowitz RA. The mannose receptor is a pattern recognition receptor involved in host defense. Curr Opin Immunol 10:50-55, 1998. 38. Sondak VK, King DW, Zager JS, et al. Combined analysis of phase III trials evaluating [99mTc] tilmanocept and vital blue dye for identification of sentinel lymph nodes in clinically node-negative cutaneous melanoma. Ann Surg Oncol 20:680-688, 2013. 39. Kampmeier O. The value of the injection method in the study of lymphatic development. Anat Rec 6:223-233, 1912. 40. Sabin FR. On the origin of the lymphatic system from the veins and the development of the lymph hearts and thoracic duct in the pig. Am J Anat 1:367-389, 1902. 41. Galland F, Karamysheva A, Pebusque MJ, et al. The FLT4 gene encodes a transmembrane tyrosine kinase related to the vascular endothelial growth factor receptor. Oncogene 8:1233-1240, 1993. 42. Jeltsch M, Kaipainen A, Joukov V, et al. Hyperplasia of lymphatic vessels in VEGF-C transgenic mice. Science 276:1423-1425, 1997. 43. Kaipainen A, Korhonen J, Pajusola K, et al. The related FLT4, FLT1, and KDR receptor tyrosine kinases show distinct expression patterns in human fetal endothelial cells. J Exp Med 178:2077-2088, 1993. 44. Oh SJ, Jeltsch MM, Birkenhager R, et al. VEGF and VEGF-C: specific induction of angiogenesis and lymphangiogenesis in the differentiated avian chorioallantoic membrane. Dev Biol 188:96-109, 1997. 45. Karkkainen MJ, Saaristo A, Jussila L, et al. A model for gene therapy of human hereditary lymphedema. Proc Natl Acad Sci U S A 98:12677-12682, 2001. 46. Buttler K, Becker J, Pukrop T, et al. Maldevelopment of dermal lymphatics in Wnt5a-knockout-mice. Dev Biol 381:365-376, 2013.

K23346_Neligan_22_Biomarkers_r3_dc_0299-0308.indd 307

5/26/2015 9:38:47 AM

K23346_Neligan_22_Biomarkers_r3_dc_0299-0308.indd 308

5/26/2015 9:38:47 AM

C hapter 23 Clinical Staging of Lymphedema Sandro Michelini, Marco Cardone, Alessandro Fiorentino

Clin

K ey P oints • There is no perfect staging system for lymphedema. • A staging system is essential to facilitate a scientific dialog and cohesive treatment planning. • In addition to staging the clinical disease, it is important to incorporate functional assessment. • Proper treatment planning can only occur with such a staging system.

There is no perfect staging system for lymphedema. This becomes patently obvious as we learn more about the condition. The present lack of a universally recognized staging system makes it difficult to compare not only diagnoses but also treatment options and outcomes in published reports. The staging of lymphedema is a long-standing question that has been discussed at consensus meetings at all national and international congresses of lymphology. To secure universal agreement about the definitions and framing of the pathology of lymphedema, a staging system characterized by simplicity, recognizability, and worldwide use is required. Four international proposals exist; they are based on different clinical aspects and instrumental measures of the pathology but have some characteristics in common. It is essential that these be synchronized and synthesized through the work of a special world commission to obtain a scientific tool with universally recognized and accepted parameters. A common tool to stage lymphedema is necessary for more precise scientific communication and medicolegal and social reasons. Particularly in its advanced stages, lymphedema is a socially significant disease with substantial cost implications for treatment and the loss of working capacity.

309

K23346_Neligan_23_Clin Staging_r3_dc_0309-0314.indd 309

5/26/2015 9:40:30 AM

310

Part IV  Diagnosis of Lymphedema

International Society of Lymphology The present staging reported in the 2013 Consensus Document of the International Society of Lymphology (ISL)1 includes one preclinical and three clinical stages (Table 23-1). The initial staging of three clinical stages was revised by the International Society of Lymphology, Lymphology Association of North America, and German Society of Lymphology. This revision underscored the importance of including a preclinical stage for both primary and secondary lymphedema, which was defined as stage 0. The three clinical stages differ essentially in the presence of a pitting (stage I) or nonpitting (stages II and III) edema and in the association in the third stage of complications, often with a progressive evolution in the skin and subcutaneous tissues. The same stages are indicated in the Consensus Document of the International Union of Phlebology.2 Stage 0 is subclinical. Stage 1 represents an early accumulation of fluid with high-protein content that subsides with limb elevation (Fig. 23-1, A). Pitting edema is seen in this stage. In stage 2, limb elevation rarely reduces tissue swelling, and pitting is not a feature (Fig. 23-1, B). Stage 3 includes lymphostatic elephantiasis with absent pitting and trophic skin changes (acanthosis, fat deposits, and warty overgrowths) (Fig. 23-1, C). The severity of the stage is based on volume dif-

TABLE 23-1  Lymphedema Staging According to the 2013 Consensus Document of the International Society of Lymphology Stage

Evidence

0

Subclinical; absence of edema in “risk of development” patient

I

Presence of edema reduced by treatment (pitting edema)

II

Edema partially reduced by treatment (no pitting edema)

III

Elephantiasis with skin lesions and relapsing infections

B

C

A

FIG. 23-1  International Union of Phlebology stages. Stage 0 (is subclinical). A, Stage 1 (pitting edema); B, stage 2 (no pitting edema); C, stage 3 (complicated elephantiasis).

K23346_Neligan_23_Clin Staging_r3_dc_0309-0314.indd 310

5/26/2015 9:40:31 AM

Chapter 23  Clinical Staging of Lymphedema

311

ferences: minimal (less than 20% increase), moderate (20% to 40% increase), and severe (greater than 40% increase). At the Twentieth International Congress of Lymphology in Brazil, various groups offered different proposals on lymphedema staging. A special international commission gathered all these ideas to define the new official staging according to the ISL.2

German Staging The German Society of Lymphology, led by Etelka Földi, included for the first time four clinical stages of lymphedema. In addition to the four stages of the ISL Consensus Document, a stage 0 was included, representing all cases of subclinical lymphedema but with significant risk of the clinical appearance of edema (for example, lymphoscintigraphic findings of lymphatic impairment)3 (Table 23-2).

Italian Staging In 1995 the Italian Society of Lymphangiology proposed five stages4 (Table 23-3), in which preclinical cases with a risk of worsening edema are underscored (stage 1). However, this school also differed in the underlying cases of elephantiasis with severe chronic inflammatory and infective complications with the risk of tumor tissue degeneration (stage 5).4-7

TABLE 23-2  Lymphedema Staging According to the German Society of Lymphology Stage

Evidence

0

No edema but significant risk of its clinical appearance

1

Edema reduced with treatment (pitting edema)

2

Edema reduced with treatment only partially (no pitting edema)

3

Elephantiasis with skin lesions and relapsing infections

TABLE 23-3  Lymphedema Staging Proposed by the Italian Society of Lymphangiology Stage

Evidence

1

No edema in an at-risk patient (preclinical)

2

Edema that reduces spontaneously with antigravitational position and during the night

3

Edema that does not reduce spontaneously and only partially with treatment; appearance of recurrent lymphangitis

4

Fibrotic edema (disappearance of tendon and bone shapes)

5

Complicated elephantiasis with relapsing skin infections and involvement of deep layers (muscles and joints)

K23346_Neligan_23_Clin Staging_r3_dc_0309-0314.indd 311

5/26/2015 9:40:31 AM

312

Part IV  Diagnosis of Lymphedema

Japanese Staging Ohkuma,8 a scientist with a particular interest in infective complications of the skin and subcutaneous tissues, proposed four stages based on clinical criteria and frequency of infections and inflammatory episodes, in which it is also possible to obtain prognostic information from this staging (Table 23-4).

South American Staging The staging system proposed by the Brazilian Society of Phlebology and Lymphology is led by the Brazilian group (Mauro Andrade). In addition to the preclinical aspects and risk of developing infectious and degenerative complications, this group stresses the functional data of the impairment resulting from the edema. According to this staging, only one major joint of the limb must be involved, although two or all three major joints can be affected. This aspect allows the establishment of a global therapeutic rehabilitative program to determine the necessity of assistance for the patient and the degree of reduction of activities of daily living9 (Table 23-5). Therefore this staging includes both clinical and functional criteria.9

TABLE 23-4  Lymphedema Staging Proposed by the Japanese School Characteristic of the Skin

Physical Examination

Lipodermatosclerosis With/Without Phlogosis

Prognosis

1

Normal

Pitting edema11

No

Temporary

2

Thin skin

Harder edema, pitting edema1

No

Permanent

3

Cutaneous lichenification

No pitting edema

Yes

Worsening edema

4

Cutaneous verrucosis

Fibrotic, no pitting edema

Very frequent

Worsening edema

Stage

1, Moderate; 11, remarkable.

TABLE 23-5  Lymphedema Staging Proposed by the Brazilian Society of Phlebology and Lymphology Stage

Evidence

0

No edema in an at-risk patient (preclinical)

1

Edema that reduces spontaneously with antigravitational position, pitting11, Stemmer sign1, and involvement of at least two joints

2

Edema that does not reduce spontaneously with antigravitational position but only by treatment, pitting1, Stemmer sign11, and involvement of at least two joints

3

Edema that reduces only partially with treatment, pitting1, Stemmer sign11, and involvement of at least three joints

4

Edema that reduces only partially with treatment, pitting1, Stemmer sign11, and involvement of at least three joints, with skin infections

1, Moderate; 11, remarkable.

K23346_Neligan_23_Clin Staging_r3_dc_0309-0314.indd 312

5/26/2015 9:40:31 AM

Chapter 23  Clinical Staging of Lymphedema

313

Conclusion The issue of lymphedema staging is important. With more precise, uniform definitions, each stage can be associated with specific levels of assistance parameters related to these pathologies, which are highly prevalent in the worldwide population. Staging is essential to guide physicians in developing an accurate diagnosis, selecting the appropriate treatment for each patient, monitoring the disease process, and determining the need for surgical intervention. In this way it is very important for primary prevention and early diagnosis to ensure the best management of this chronic illness. Early treatment and clinical management provide the best quality of life for the patient and less costs for both the individual and society. Currently, only the health care systems in some states and a few regions support lymphedema patients. Frequently the support available is inadequate, resources are poor, and patients are required to pay for their treatments and support systems. Therefore it is imperative to devise a common scientific tool for staging to facilitate a discussion of these problems at the regional, national, and international levels, as well as to promote epidemiologic studies on the serious disabilities caused by lymphedema. In recent years more centers around the world have started to apply the International Classification of Functioning, Disability and Health (ICIDH),10-12 because this organization provides a common framework for describing health conditions and outcome measurements. In addition to including the staging systems currently used, obtaining a disability quantification through an evaluation of specific core sets makes it possible to better describe the health condition in those patients, thereby providing a more complete tool for clinical evaluation and treatment planning.13-15

C linical P earls • Stemmer’s test results in either a positive or negative sign for primary lymphedema in all stages. • Pitting test is usually present in the early stages of lymphedema. • Fibrosis and elephantiasis are common in the advanced stages. • International Classification of Functioning, Disability and Health (ICF) is strongly recommended for describing disability and functional status.

R EFERENCES 1. International Society of Lymphology. The diagnosis and treatment of peripheral lymphedema: 2013 Consensus Document of the International Society of Lymphology. Lymphology 46:1-11, 2013. 2. Lee B, Andrade M, Bergan J, et al. Diagnosis and treatment of primary lymphedema. Consensus Document of the International Union of Phlebology (IUP)-2009. Int Angiol 29:454-470, 2010.

K23346_Neligan_23_Clin Staging_r3_dc_0309-0314.indd 313

5/26/2015 9:40:31 AM

314

Part IV  Diagnosis of Lymphedema

3. Földi M, Földi E. Földi’s Textbook of Lymphology. New York: Elsevier, 2009. 4. Campisi C, Michelini S, Boccardo F, et al. Modern stadiation of lymphedema and corresponding preventive options. Eur J Lymphol 7:27-31, 1999. 5. Michelini S, Campisi C, Gasbarro V, et al. National guidelines on lymphedema. Lymphology 55:238242, 2007. 6. Michelini S, Failla A, Moneta G, et al. Clinical staging of lymphedema and therapeutical implications. Lymphology 35:168-176, 2002. 7. Michelini S, Failla A. Linfedemi: inquadramento diagnostico clinico e strumentale. Minerva Cardioangiol 45(Suppl 1):11-15, 1997. 8. Twentieth International Congress of International Society of Lymphology [abstract]. I Brasilian Congress of Lymphology—I Congreso del CAPAL (Latin American Charter of International Society of Lymphology), Salvadore, Brazil, 2005. 9. World Health Organization. International Classification of Functioning, Disability and Health (ICF). Geneva, Switzerland: WHO, 2002. 10. Michelini S, Failla A, Moneta G, et al. International classification of lymphedema functioning and disability evaluation. Eur J Lymphol 17:16-19, 2007. 11. International Lymph Framework: Best Practice for the Management of Lymphoedema, ed 2, 2012. Available at www.lympho.org. 12. Devoogdt N. The lymphoedema functioning disability and health questionnaire for lower limb lymphoedema (Lymph-ICF-LL): reliability and validity [abstract]. Twenty-fourth Congress of the International Society of Lymphology, Rome, Sept 2013. 13. Viehoff P. Developing of ICF core sets for lymphoedema: qualitative research [abstract]. Twenty-fourth Congress of the International Society of Lymphology, Rome, Sept 2013. 14. Hendrickx AA. The use of clinimetric instruments according to the International Classification of Functioning, Disability and Health in a multidisciplinary setting [abstract]. Twenty-fourth Congress of the International Society of Lymphology, Rome, Sept 2013. 15. Michelini S, Campisi C, Failla A, et al. Staging of lymphedema: comparing different proposals. Eur J Lymphol 16:7-10, 2006.

K23346_Neligan_23_Clin Staging_r3_dc_0309-0314.indd 314

5/26/2015 9:40:31 AM

C hapter 24 Measuring Methods Peter C. Neligan

Me

K ey P oints • No perfect measurement tool for lymphedema exists. • Limb circumference and volume are the most commonly performed measurements for lymphedema. • Volume is most frequently calculated using the truncated cone model. • Perometry is a reliable method of measuring limb volume, but it is not universally available. • Lymph flow measurement using lymphoscintigraphy is a reliable way to document lymphedema. • MRI provides excellent information but is not easily applicable for documenting ongoing changes. • Bioimpedance analysis is probably the most useful method for diagnosing preclinical lymphedema.

Making a diagnosis is one of the most important things that physicians do when they first see a patient with lymphedema. Evidence suggests that a subjective assessment through patient selfreporting is a more sensitive and less expensive means of diagnosing the development of lymphedema.1-3 Nevertheless, it is useful to maintain objective documentation. A combination of both approaches is probably best. The first step in diagnosing lymphedema is to determine the extent of the disease. This requires some sort of measurement. A means of comparative and quantitative assessment of the results of treatment is also required. The measurement technique needs to be standardized, quantitative, repeatable, and reproducible; it should also be easily available and inexpensive. The most often used quantitative assessment of lymphedema is assessment of size based on the measurement of circumference or volume. Other methods include measurements of lymph flow, tonometry (to evaluate compressibility), and bioimpedance.4-6 Each of these methods has advantages and disadvantages. No perfect measuring technique exists.

315

K23346_Neligan_24_Measuring_r3_0315-0326.indd 315

5/26/2015 9:42:33 AM

316

Part IV  Diagnosis of Lymphedema

Measurement of Limb Circumference and Limb Volume Limb circumference and limb volume are the most frequently obtained measurements in patients with lymphedema.

Measurement of Limb Circumference Limb circumference is measured from a fixed point such as the tip of the long finger or from anatomic landmarks such as the patella or the olecranon (Fig. 24-1). Identification of local landmarks can be problematic in lymphedematous patients, because they may not be readily palpable. Furthermore, landmark descriptions may have various interpretations, which can lead to confusion and inaccuracy. For example, the patella can be measured from the proximal end, the distal end, or the middle. This can make a significant difference, particularly when measurements are repeated and compared at different time points. Limb circumference as a means of measurement has been criticized because of inconsistencies in tape measure tension and placement from one measurement to the next. Although measurements at specified intervals from a fixed point may seem to be a more accurate technique, Taylor et al7 found excellent correlation between measurements taken in reference to fixed anatomic landmarks and plethysmography.

FIG. 24-1  A tape measure is used to document limb circumference measurements at 4 cm intervals from the tip of the second toe. Each point is marked, and limb circumference measurements are recorded at each point of measurement.

K23346_Neligan_24_Measuring_r3_0315-0326.indd 316

5/26/2015 9:42:33 AM

Chapter 24  Measuring Methods

317

FIG. 24-2  Water-displacement plethysmography is an accurate way of measuring limb volume. The arm (or leg) is immersed in a container of water. The volume of displaced water is equivalent to the volume of the limb.

Measurement of Limb Volume The most widely accepted measure of lymphedema is limb volume. The affected limb can be compared with the unaffected limb, or measurements of the affected limb can be compared before and after interventions or events that led to the lymphedema. Volumes are most accurately measured by plethysmography or water displacement8-10 (Fig. 24-2). However, defining and replicating the upper level for immersion can be difficult, and water displacement is not convenient for routine clinical use. Brorson and Höijer11 followed 10 women with unilateral upper extremity lymphedema after breast cancer treatment. They calculated volume indirectly by circumference measurements (CM), using the formula for a truncated cone. The authors developed two Excel-based formulas of the truncated cone: one for fixed 4 cm intervals, leading to 10 volume segments (VS) (CM-10-VS), and one for varying intervals, leading to four volume segments (CM-4-VS). Plethysmography yielded greater volumes, because the hand was included, but excellent correlation was observed between volumes measured by plethysmography and volumes measured by limb circumference from a fixed point. No difference was noted between CM-10-VS and CM-4-VS. In my practice I have adopted the CM-10-VS method, because in my experience, it is simple, reproducible, and repeatable, regardless of who obtains the measurements. For the upper limb, circumference is measured at 4 cm intervals from the tip of the long finger, and for the lower limb, similar measurements are taken at 4 cm intervals from the tip of the second toe. A Perometer (Pero-System) is an optoelectronic imaging device designed to measure the volume of an object. It is ideally suited for measuring limb volume. It consists of a square measuring frame that contains rows of infrared light–emitting diodes on two adjacent sides and rows

K23346_Neligan_24_Measuring_r3_0315-0326.indd 317

5/26/2015 9:42:34 AM

318

Part IV  Diagnosis of Lymphedema

FIG. 24-3  Inside a Perometer, two rows of measurement arrays are placed at a 90-degree angle to each other. Each determines an object diameter and its position inside the frame.

of corresponding sensors on the other two sides12 (Fig. 24-3). Two rows of measurement arrays are placed at a 90-degree angle to each other. Each of them determines an object diameter and its position inside the frame. The patient sits at one end of the Perometer, with the hand or foot resting centrally on an adjustable support or footplate. The frame is then moved along the length of the arm or leg, from the wrist to the axilla or from the ankle to the thigh. When the frame is moved manually, the diameters and positions are determined in short distances along the object. The collected data are not influenced by the position of the measured object within the frame. A computer produces a volume picture of the entire limb by using the cross-sectional information obtained from the biplanar shadow of the limb in the device. The collected data can be saved and analyzed. Data generated by a Perometer can be exported into other programs such as word processing programs, spreadsheets, or other standard software. Perometry has been rigorously assessed by comparison with other methods and is considered more accurate than tape measurements.12,13 In various studies it has proved highly reproducible, accurate, and reliable.14 This technology is safe and fulfills many of the criteria for an ideal measurement tool. However, it is not inexpensive or easily accessible.

Nuances of Circumference and Volume Measurements One of the biggest confounders of circumference and volume measurements is that causes of increased volume other than lymphedema are not taken into account. For example, breast cancer

K23346_Neligan_24_Measuring_r3_0315-0326.indd 318

5/26/2015 9:42:34 AM

Chapter 24  Measuring Methods

319

patients tend to gain weight after having chemotherapy.15 Measurement of changes in limb circumference or limb volume measurements alone is not helpful in distinguishing between increases from lymphedema and other causes. This confounder can be somewhat controlled in patients with unilateral lymphedema by comparing the affected side with the contralateral, normal limb. In patients with bilateral lymphedema, however, this method is not as useful.

Measurement of Lymph Flow One useful diagnostic criterion for diagnosing lymphedema is the documentation of evidence of slow, reduced, or completely absent lymphatic flow in the swollen tissue. Lymphoscintigraphy is generally considered to be the benchmark procedure for measuring flow. It involves the injection of technetium-99m–labeled, filtered sulfur colloid into the subcutis in the web spaces of the fingers or toes. The labeled colloid is taken up by the lymphatics and transported to the draining nodal basin (Fig. 24-4). The flow can be detected and visualized with scintigraphic scanning. A transport index (TI) score can be calculated to categorize the lymphatic flow as normal or pathologic. This takes several factors into account, including lymphatic transport kinetics, the distribution pattern, and the appearance time of lymph nodes, and provides assessment data of lymph nodes at two time-points (Table 24-1). A score of less than 10 signifies a normal transport index. Scores are reported bilaterally, even in cases of unilateral swelling.16

FIG. 24-4  Lymphoscintigram of a swollen right leg with impaired lymphatic clearance, indicated by the accumulation of isotope in the lower leg.

K23346_Neligan_24_Measuring_r3_0315-0326.indd 319

5/26/2015 9:42:34 AM

320

Part IV  Diagnosis of Lymphedema

TABLE 24-1  Components of Transport Index Score

0

3

5

9

Transport kinetics

No delay

Mild delay

Extreme delay

No flow

Distribution patterns

Normal

Partial dermal

Diffuse dermal

No flow

Time index

Time in minutes for appearance of regional lymph nodes, multiplied by 0.04

Lymph nodes

Normal

Visible, diminished

Barely visible

Not seen

Lymph vessels

Normal

Visible, diminished

Barely visible

Not seen

A normal transport index score is less than 10.

A

B

FIG. 24-5  MR lymphangiograms in two patients with lymphedema. A, Limited subdermal lymphatics. B, Abundant subdermal lymphatics (arrows).

The advantage of this index, with five different characteristics of the lymphatic system, is that it is helpful for identifying the varied disease processes that can be present within this system. In a prospective study of 386 extremities, Cambria et al17 identified patients in whom only the distribution pattern was abnormal; the transport kinetics were within normal limits. In other patients, the transport was delayed but the distribution pattern was normal. For this reason, the use of all five components of the transport index adds not only to its utility in diagnosing lymphedema, but also to its usefulness for defining specific anatomic details of the lymphatic system in each patient. More recently, MR lymphangiography (see Chapter 28) has been used to obtain good information about flow and therefore lymphatic function. It also provides high-definition images of the location and pattern of subdermal lymphatics (Fig. 24-5). This is invaluable information for plan-

K23346_Neligan_24_Measuring_r3_0315-0326.indd 320

5/26/2015 9:42:35 AM

Chapter 24  Measuring Methods

321

ning lymphatic surgery. Furthermore, information gathered from MR images can be used to very accurately measure limb volume. The amount and location of edema in the limb can be documented using this technology. However, this is an expensive, cumbersome, labor-intensive, and time-consuming method of limb volume measurement that is not universally available.

Tonometry Tissue tonometry is an easy-to-use, fast, accurate, and reproducible method of assessing pitting edema and fibrotic changes in edematous tissue. Pitting edema is present in stage I (International Society of Lymphology [ISL]) lymphedema and is the result of displacement of extracellular fluid (Fig. 24-6). With the progression of lymphedema, chronic inflammatory changes occur that result in progressive fibrotic induration. These are seen in patients with ISL stage II lymphedema. A tonometer is a mechanical device that pushes a plunger into the skin (Fig. 24-7). The depth of penetration of the plunger is recorded on a dial to a resolution of 0.01 mm. In early stage lymphedema, when pitting is present, the plunger can be pushed deeper into the skin. As the condition

FIG. 24-6  Finger pressure demonstrates pitting edema in this patient. The indentation remains after the finger is removed.

FIG. 24-7  A mechanical tonometer consists of a plunger that is pressed onto the skin and a scale for measuring the depth of penetration.

K23346_Neligan_24_Measuring_r3_0315-0326.indd 321

5/26/2015 9:42:35 AM

322

Part IV  Diagnosis of Lymphedema

progresses and fibrosis becomes more prominent, the degree of penetration decreases.18 Assessment of the rate of this change is useful for determining the impact of treatment and management of fibrosis in the patient, which is a major cause of poor lymph drainage and poor treatment outcomes.

Bioimpedance Spectroscopy Bioimpedance spectroscopy (BIS) analysis is a technology that uses resistance to electrical currents in comparing the composition of fluid compartments within the body. It is one of several techniques that directly measures lymph fluid volume. BIS is based on the theory that the amount of resistance to the flow of electrical current (impedance) through the body is inversely proportional to the volume of fluid in the tissue. In lymphedema, tissue impedance decreases with the accumulation of excess interstitial fluid. In BIS, an electrical current is passed through a body segment, and impedance to the flow of the current is measured. It operates on the premise that tissues such as fat and bone act as insulators, while electrolytic fluids conduct electricity. The properties unique to lymphatic fluid can be determined based on the measurements of the current flow. Low-frequency currents selectively pass through extracellular fluid compartments, whereas high-frequency currents pass through both intracellular and extracellular fluid. Although BIS can accurately measure extracellular accumulation of lymphatic fluid, it is not useful for quantifying other tissue elements that increase, such as fibrous and adipose tissue. Its utility in the measurement of chronic lymphedema characterized by increased fibrosis and fat deposition is therefore questionable. To determine the optimal frequency for bioimpedance measurement, Cornish et al19 measured limb impedance at 256 frequencies ranging from 3 kHz to 1000 kHz for a sample control population and in patients with arm lymphedema and leg lymphedema. They concluded that impedance measurements higher than 30 kHz decrease sensitivity to extracellular fluid and are not reliable for early detection of lymphedema. With the use of BIS, the affected limb and the contralateral unaffected limb can be compared. The measured impedance of both limbs can then be expressed as a ratio. Because impedance declines as extracellular fluid volume increases, the measured bioimpedance values are usually expressed as a ratio of the normal limb to the abnormal limb. In a person without lymphedema, this ratio should be 1. However, as the severity of lymphedema increases, the ratio rises proportionately. The presence of bilateral lymphedema poses particular challenges with regard to the use of BIS. As discussed previously, unilateral lymphedema can be monitored and quantitated through the use of the contralateral, normal limb as a reference with which to construct the bioimpedance ratio. For the assessment of bilateral lymphedema, the quantitation of intracellular fluid volume can serve as a suitable alternative reference.20 In theory, because intracellular fluid volume should be virtually unaffected by the onset or progression of lymphedema, an extracellular/intracellular fluid volume ratio can be constructed from the measured BIS.

K23346_Neligan_24_Measuring_r3_0315-0326.indd 322

5/26/2015 9:42:35 AM

Chapter 24  Measuring Methods

323

In prospective evaluations to date, patients and practitioners have found BIS to be rapid, accurate, and consistent. It is particularly useful for detecting preclinical lymphedema. In one study with BIS, lymphedema was detected 10 months before the condition could be clinically diagnosed.19 This may be the main strength of this technology; evidence shows that early treatment of lymphedema improves outcomes.21 Despite much evidence to support the efficacy of BIS, some authors have reported otherwise. Ward et al20 assessed the agreement between bioimpedance indices and interlimb volume differences. They used perometry to assess unilateral arm lymphedema. Despite a strong linear correlation between bioimpedance and perometric measurements (r 5 0.919), bioimpedance gave falsenegative results for 12 (27%) patients who had lymphedema. Box et al22 investigated the incidence of arm lymphedema after axillary dissection to determine the effect of prospective monitoring and early physiotherapy intervention. They used three measurements to detect arm lymphedema: arm circumferences (CIRC), arm volume (VOL), and multifrequency bioimpedance (MFBIA). An increase of at least 200 ml over the preoperative difference between the two arms indicated clinically significant lymphedema. The CIRC and MFBIA methods failed to detect lymphedema in up to 50% of women with an increased volume of at least 200 ml in the operated arm, compared with the unoperated arm. Fu et al23 examined the reliability, sensitivity, and specificity of BIS in the detection of lymphedema in 250 women. Some were healthy, some were breast cancer survivors with lymphedema, and others were at risk for developing lymphedema. They used circumferential tape measurements to validate the presence of lymphedema. Lymph fluid changes were measured with bioimpedance. The bioelectrical impedance analysis ratio, as indicated by an L-Dex ratio, was highly reliable in healthy women and survivors at risk for lymphedema. Reliability was acceptable for survivors with lymphedema. The bioelectrical impedance ratio correlated with limb volume assessed by sequential circumferential tape measurement. However, the L-Dex ratio had a diagnostic cutoff of greater than 17; it is thought that the 20% of lymphedema cases that are potentially missed is a problem related to this particular device and not necessarily to the technology. The authors noted the importance of integrating other assessment methods, such as self-reporting, clinical observation, or perometry, to ensure the accurate detection of lymphedema. The effectiveness of BIS is controversial in the literature. In the United States, most insurers consider it investigational.

C linical P earls • When limb circumference is consistently measured from a fixed point, it is a very effective means of determining limb volume. • MR lymphangiography provides accurate measurements of limb volume but is expensive, time consuming, and not readily available to many physicians. • Information obtained with a transport index is useful for defining unique anatomic details of the lymphatic system in each patient. • Lymphedema should be diagnosed and quantitated using a variety of assessment tools.

K23346_Neligan_24_Measuring_r3_0315-0326.indd 323

5/26/2015 9:42:35 AM

324

Part IV  Diagnosis of Lymphedema

R EFERENCES 1. Armer JM, Radina ME, Porock D, et al. Predicting breast cancer-related lymphedema using selfreported symptoms. Nurs Res 52:370-379, 2003. 2. Gartner R, Jensen MB, Kronborg L, et al. Self-reported arm-lymphedema and functional impairment after breast cancer treatment: a nationwide study of prevalence and associated factors. Breast 19:506515, 2010. 3. Armer JM, Stewart BR. A comparison of four diagnostic criteria for lymphedema in a post-breast cancer population. Lymphat Res Biol 3:208-217, 2005. 4. Armer JM. The problem of post-breast cancer lymphedema: impact and measurement issues. Cancer Invest 23:76-83, 2005. 5. Gerber LH. A review of measures of lymphedema. Cancer 83:2803-2804, 1998. 6. Ridner SH, Montgomery LD, Hepworth JT, et al. Comparison of upper limb volume measurement techniques and arm symptoms between healthy volunteers and individuals with known lymphedema. Lymphology 40:35-46, 2007. 7. Taylor R, Jayasinghe UW, Koelmeyer L, et al. Reliability and validity of arm volume measurements for assessment of lymphedema. Phys Ther 86:205-214, 2006. 8. Petrek JA, Pressman PI, Smith RA. Lymphedema: current issues in research and management. CA Cancer J Clin 50:292-307, 2000. 9. Megens AM, Harris SR, Kim-Sing C, et al. Measurement of upper extremity volume in women after axillary dissection for breast cancer. Arch Phys Med Rehab 82:1639-1644, 2001. 10. Kaulesar Sukul DM, den Hoed PT, Johannes EJ, et al. Direct and indirect methods for the quantification of leg volume: comparison between water displacement volumetry, the disk model method and the frustum sign model method, using the correlation coefficient and the limits of agreement. J Biomed Eng 15:477-480, 1993. 11. Brorson H, Höijer P. Standardised measurements used to order compression garments can be used to calculate arm volumes to evaluate lymphoedema treatment. J Plast Surg Hand Surg 46:410-415, 2012. 12. Stanton AW, Northfield JW, Holroyd B, et al. Validation of an optoelectronic limb volumeter (Perometer). Lymphology 30:77-97, 1997. 13. Tierney S, Aslam M, Rennie K, et al. Infrared optoelectronic volumetry, the ideal way to measure limb volume. Eur J Vasc Endovasc Surg 12:412-417, 1996. 14. Moseley A, Piller N, Carati C. Combined opto-electronic perometry and bioimpedance to measure objectively the effectiveness of a new treatment intervention for chronic secondary leg lymphedema. Lymphology 35:136-143, 2002. 15. Lankester KJ, Phillips JE, Lawton PA. Weight gain during adjuvant and neoadjuvant chemotherapy for breast cancer: an audit of 100 women receiving FEC or CMF chemotherapy. Clin Oncol 14:64-67, 2002. 16. Kleinhans E, Baumeister RG, Hahn D, et al. Evaluation of transport kinetics in lymphoscintigraphy: follow-up study in patients with transplanted lymphatic vessels. Eur J Nucl Med 10:349-352, 1985. 17. Cambria RA, Gloviczki P, Naessens JM, et al. Noninvasive evaluation of the lymphatic system with lymphoscintigraphy: a prospective, semiquantitative analysis in 386 extremities. J Vasc Surg 18:773782, 1993. 18. Piller NB, Clodius L. The use of a tissue tonometer as a diagnostic aid in extremity lymphoedema: a determination of its conservative treatment with benzo-pyrones. Lymphology 9:127-132, 1976.

K23346_Neligan_24_Measuring_r3_0315-0326.indd 324

5/26/2015 9:42:35 AM

Chapter 24  Measuring Methods

325

19. Cornish BH, Chapman M, Hirst C, et al. Early diagnosis of lymphedema using multiple frequency bioimpedance. Lymphology 34:2-11, 2001. 20. Ward LC, Essex T, Cornish BH. Determination of Cole parameters in multiple frequency bioelectrical impedance analysis using only the measurement of impedances. Physiol Meas 27:839-850, 2006. 21. Boccardo FM, Ansaldi F, Bellini C, et al. Prospective evaluation of a prevention protocol for lymphedema following surgery for breast cancer. Lymphology 42:1-9, 2009. 22. Box RC, Reul-Hirche HM, Bullock-Saxton JE, et al. Physiotherapy after breast cancer surgery: results of a randomised controlled study to minimise lymphoedema. Breast Cancer Res Treat 75:51-64, 2002. 23. Fu MR, Cleland CM, Guth AA, et al. L-Dex ratio in detecting breast cancer-related lymphedema: reliability, sensitivity, and specificity. Lymphology 46:85-96, 2013.

K23346_Neligan_24_Measuring_r3_0315-0326.indd 325

5/26/2015 9:42:35 AM

K23346_Neligan_24_Measuring_r3_0315-0326.indd 326

5/26/2015 9:42:35 AM

C hapter 25 Hydromechanics of Intercellular Fluid and Lymph Waldemar Olszewski, Marzanna T. Zaleska

Hyd K ey P oints • In normal limbs, lymph flow occurs only during spontaneous contractions of lymphangions. • In obstructive lymphedema, the stagnant tissue fluid is primarily deep in the subcutaneous tissue. • In advanced stages of lymphedema, the hydraulic conductivity of the subcutis decreases. • Tissue fluid pressure is low in advanced stages of lymphedema because the interstitial spaces of the subcutis expand. • Compression pressures of 50 to 60 mm Hg are recommended in early stages of lymphedema, and pressures of up to 125 mm Hg are needed in advanced stages.

Human limb soft tissues are composed of skin; subcutaneous tissue containing loose connective tissue structures such as ground matrix, fibers, and adipocytes; nerve fibers; blood and lymphatic vessels; muscular fascia; and muscle fibers. All of these tissue elements bathe in the fluid filtered from blood exchange vessels, which is partly mobile and partly taken up by cells bound to the matrix. In cases of enhanced plasma filtration or obstruction of tissue fluid flow to the lymphatics, and further, to the blood circulation, the interstitial space becomes overloaded with fluid. This is clinically diagnosed as tissue edema. In the lower limbs, the estimated extravascular extracellular volume is 12% of the total tissue volume under normal conditions, whereas in lymphatic obstruction it may be 40% to 50%.1 Knowledge of the location of excess fluid and its pressure/flow mechanics is necessary to understand the role of tissue fluid in the metabolism of parenchymal cells under normal conditions and in patients with lymphedema from obstruction of lymph flow. Such information is indispensable for effective antiedema therapy.

327

K23346_Neligan_25_Hydromechanics_r2_dc_0327-0048.indd 327

5/26/2015 9:46:35 AM

328

Part IV  Diagnosis of Lymphedema

Various external forces in specific directions should be applied in compression therapy, depending on the mobile fluid topography at different limb levels; the fluid location in the deep layers of the subcutaneous tissue, around blood vessels and nerves, and above and under muscular fascia; and the hydraulic conductivity at various levels.

Accumulation of Tissue Fluid and Lymph Under Normal Conditions and in Obstructive Lymphedema Our understanding of the limb lymphatic network in physiologic conditions and in lymphedema is based on lymphographic or lymphoscintigraphic images of the superficial and deep systems and lymph nodes. This technique does not reveal minor lymphatic structures located under the epidermis. Direct lymphangiography with fluorescent tracers may be helpful in delineating minor dermal lymphatics but is rarely used, as it requires special equipment for visualization. Ultrasonography, CT, and MRI provide pictures of tissue spaces filled with stagnant tissue fluid; however, they do not show lymphatics. None of the listed methods provides sufficient imaging of the entire tissue fluid and lymph space—including the interstitial space and lymphatics—to know what these tissues look like in reality. It is difficult to imagine how tissue fluid in the areas with obstructed lymphatics finds its way to the normal, noncongested tissue regions and is absorbed. To date, it is only through anatomic dissection and histologic processing of biopsy material that one can visualize the tissue lymphatic network and the sites of accumulation of the excess mobile tissue fluid.1 Under normal conditions, the volume of mobile tissue fluid is negligible. Collecting lymphatics contain little lymph in the lymphangions (segments of lymphatic vessels bounded by valves). Some of them remain empty. The situation changes dramatically with the obstruction of lymph flow caused by damage to the lymph collectors: subepidermal lymphatics dilate and tissue fluid spaces form in the subcutaneous tissue around small veins and in the muscular fascia. The most superficial layer where fluid accumulates is the subepidermal lymphatic plexus, occupying a papillary and reticular dermis with a thickness of 200 to 300 microns (Fig. 25-1). However, the volume of fluid in this plexus is negligible, compared with the volume of the subcutaneous tissue fluid, and does not exceed 2% to 3% of the total tissue fluid retained in soft tissues. The bulk (95%) of mobile tissue fluid accumulates in the subcutaneous tissue, forming artificial, partly interconnected spaces (Fig. 25-2). These spaces are located between fat globules, fibrous bundles, and around small veins. The formation of large lakes of tissue fluid can be explained by the presence of lax connective tissue in these regions, its high compliance and subsequently low resistance to fluid flow. Tissue fluid channels form, which mostly are narrow longitudinal spaces between the normal fascial fibrous elements around and in the hypertrophic muscular fascia of the calf, sometimes reaching 2 cm in diameter. The hydraulic conductivity of these structures is high because of the linear positioning of fibers.2 The volume of fluid accumulating in the tissue spaces and calculated from the densitometric readings of the stained tissues can be as high as 40% to 50% of the total tissue volume1

K23346_Neligan_25_Hydromechanics_r2_dc_0327-0048.indd 328

5/26/2015 9:46:35 AM

Chapter 25  Hydromechanics of Intercellular Fluid and Lymph

329

A

B

C

FIG. 25-1  Histologic image of skin and subcutaneous tissue in advanced lymphedema. A, The arrow points to multiple dilated (blue stained) subepidermal lymphatic vessels. B, Magnification of blue-stained subepidermal lymphatic vessels. C, The arrow points to dilated spaces between collagen bundles filled with excess tissue fluid (H&E, 2003).

A

B

FIG. 25-2  Histologic image of the subcutaneous tissue in advanced lymphedema. A, Collagen bundles are separated by tissue fluid. B, Tissue spaces are stained blue after intratissue injection with patent blue dye. The walls of the spaces are not lined by lymphatic endothelial cells. The area occupied by excess fluid is 40% to 50% of the total tissue (H&E, 6003).

K23346_Neligan_25_Hydromechanics_r2_dc_0327-0048.indd 329

5/26/2015 9:46:36 AM

330

Part IV  Diagnosis of Lymphedema

A

Epidermis Dermis Superficial lymphatic plexis

B

Subcutis No lymphatics Tissue spaces Fascia Perimuscular space

FIG. 25-3  A, Fluid distribution in the skin and subcutaneous tissue in lymphedema. Dilated subepidermal lymphatics (upper arrow) are evident below the epidermis. The subcutaneous tissue contains no lymphatics. The bulk of mobile fluid is in the subcutis, deep at the muscular fascia (center arrow), and sometimes in the muscular compartment (lower arrow). B, This skin and subcutaneous tissue specimen was removed during a debulking operation in a patient with stage IV lymphedema. The vertical arrow shows the thickness of the tissue with the most mobile tissue fluid, compared with the dermis. The horizontal arrow points to the level at which external compression should be applied to mobilize fluid.

(Fig. 25-3, A). Anatomic images of the skin and subcutis in lower and upper limb lymphedema show that the thickness of edematous tissues is 1 to 10 cm or more (Fig. 25-3, B). (The staging system referred to in this chapter is that of W.L. Olszewski.14) This information raises the following question: How much massaging force should be applied to affect the deepest edematous tissue layers? In this chapter, we attempt to answer this question.

Tissue Resistance to Flow and Dissipation of External Force Which tissue elements create tissue resistance to fluid flow, and where is most of the external force dissipated? The answers to these questions are presented in Fig. 25-4. Skin has inherent stiffness and becomes even harder in advanced stages of lymphedema because of continuing fibrosis. The subcutaneous tissue contains collagen, elastic fibers, and fat agglomerates. Its stiffness is lower than that of skin. However, in lymphedema, fibrogenesis makes it less elastic. These structures create resistance to flow. To move mobile tissue fluid, a pressure gradient should be generated between the collagen bundles and the ground matrix, perpendicular to the limb axis (middle horizontal arrow). The hydraulic conductivity of the subcutis thus steadily decreases. Higher external pressure will be needed to mobilize edema fluid in the deeper layers. The subcutaneous tissue becomes swollen and is relatively thicker than the skin.

K23346_Neligan_25_Hydromechanics_r2_dc_0327-0048.indd 330

5/26/2015 9:46:36 AM

Chapter 25  Hydromechanics of Intercellular Fluid and Lymph

Subcutis Epidermis

331

Fascia Muscles

FIG. 25-4  Tissue sites where applied external force is dissipated. The horizontal arrows and the vertical arrow pointing upward represent the direction of applied force. The ground substance (white space in the subcutis), the pores between fibers (green dots), and the collagen bundles (brown lines) create resistance to flow, represented by the arrows pointing downward.

The amount of force lost during external compression can be estimated by measuring the pressure of the therapist’s hand on the skin, or measuring the undergarment pressure of a bandage or pneumatic device on the skin, and comparing that with tissue fluid pressure. The difference between applied and in-tissue, generated pressure will provide information on how much force was absorbed by the solid tissue and how much was used for generating the fluid pressure gradient necessary for propelling tissue fluid. Thus loss of force should always be taken into account when planning compression procedures. Compression-generated tissue fluid pressures are always lower than pressures applied at the compression device–skin interface.3

Measurement Techniques Intralymphatic Pressure and Flow To measure intralymphatic lymph pressure, a subcutaneous lymph vessel of the leg is cannulated against the direction of lymph flow, according to published techniques.4,5 The lymph vessel draining lymph from the skin, subcutaneous tissue, and perimuscular fascia of the foot and anterior aspect of the lower leg is cannulated. Pressures are recorded by an electronic micromanometer. To measure intralymphatic flow, we use a low-flow flowmeter that measures from 0.1 to 6 ml/min, with an accuracy of 0.1 ml.5

K23346_Neligan_25_Hydromechanics_r2_dc_0327-0048.indd 331

5/26/2015 9:46:37 AM

332

Part IV  Diagnosis of Lymphedema

Tissue Fluid Pressure and Flow To measure tissue fluid pressure, the wick-in-needle technique is routinely used. An 8-gauge injection needle with polyethylene tubing (outer diameter 1.34 mm) containing a glass-wool wick protruding 5 mm from the tubing tip is introduced under the skin. The outer part of the tubing is connected to the pressure transducer, and recordings are obtained using a three- or six-channel device. We use LabVIEW software.3 Tissue fluid flow is measured using strain-gauge plethysmography. The device measures circumference changes in the calf and thigh segments corresponding to the compressed limb region or sequentially inflated pneumatic sleeve chambers. The data are used for calculating volume changes of the limb caused by the proximally moved tissue fluid. The volume value obtained before compression is subtracted from that obtained during compression, providing data on the proximally transferred fluid volume.3

Characteristics of Lymph and Tissue Fluid Normal Tissues Lymph and tissue fluid hydraulics in human limb soft tissues, under normal conditions and in lymphedema, have been studied only in a few centers.6-11 The hydromechanics of lymph and mobile tissue fluid (normal and stagnant edema) differ considerably. Lymph is contained in the lumen of a rhythmically contracting vessel generating a pressure gradient, whereas tissue fluid is contained in the interstitial space, limited by cells and fibers. (Hydraulic conditions change dramatically with obstruction of the draining lymphatics.)

Extrinsic Factors Propelling Lymph Muscular activity, respiratory movements, passive movements, and arterial pulsation have no effect on lymph flow.6-8,10,11 Generally, limb lymphatics are empty, with only a few microliters of lymph in some lymphangions. In a normal leg positioned upright, the lymphatics have no hydrostatic pressure.8,11

Intrinsic Factors Propelling Lymph Lymph is propelled by autonomous rhythmic contractions of lymphangions.6-11 Tissue fluid enters the initial lymphatics to flow into the lymphangions. Stretching of lymphatic walls by inflowing tissue fluid causes contractions of the lymphatic wall muscles (according to Starling’s law for the heart muscle) and generates flow.

K23346_Neligan_25_Hydromechanics_r2_dc_0327-0048.indd 332

5/26/2015 9:46:37 AM

333

Chapter 25  Hydromechanics of Intercellular Fluid and Lymph

Lymph Pressures Limb lymphatics contract rhythmically; the frequency depends on the volume of inflowing tissue fluid (Fig. 25-5). The frequency is high in regions with a high capillary filtration rate and tissue fluid formation. Table 25-1 outlines normal lymph pressures. Massaging of the foot or tapping of tissues containing lymphatics has no effect on lymph pressures. Heating of the foot significantly increases the pressure, amplitude, and frequency of lymphatic contractions.8,11

1 min

ml (1 ml 5 1 mm) 50 40 30 20 5 0

15

mm Hg

10

5 0 Patient supine: no movement

FIG. 25-5  Pressure (lateral) and flow recorded in a normal calf lymphatic vessel. Three pulse waves (red curves) are shown. Each has a different amplitude, and the time intervals vary between contractions. Each lymphangion contraction generates pressures, propelling flow (blue curve). The ascending fragment of each curve shows the stroke volume. Flow occurred only during lymphangion contractions.

TABLE 25-1  Lymph Hydraulics in Normal Leg Lymphatic Collectors

Free Proximal Flow (lateral pressure) Upright

Lying Down

Foot Flexing

Pressure (mm Hg)

7-30

7-30

10-30

Pulse amplitude (mm Hg)

5-17

Pulse frequency (pulses/min)

2-8

K23346_Neligan_25_Hydromechanics_r2_dc_0327-0048.indd 333

Obstructed Flow in Normal Lymphatics (mimicking lymphatic obstruction in postsurgical lymphedema) (end pressure) Lying Down

Foot Flexing

15-55

15-50

3-20

3-35

3-14

0.6 to 6

2.5-10

3-12

5/26/2015 9:46:37 AM

334

Part IV  Diagnosis of Lymphedema

15

mm Hg

10

5

0 Patient 1 supine: no movement

Patient 1 supine: movement

Patient 2 supine: no movement

FIG. 25-6  Lymph pressure recorded in two patients with stage III lymphedema. Spontaneous pressure waves generated by damaged lymphangions are low (flat blue lines) and unable to create flow.

Lymphatic Obstruction In patients with postinflammatory, postoperative, posttraumatic, or idiopathic lymphedema, intralymphatic pressures and flow are abnormal because of (1) destruction of lymph vessel muscle cells, (2) destruction of valves, and (3) partial or total obstruction of the lumen. Tissue fluid flows to the nonswollen parts of the limb, along the hydraulically created tissue channels.

Lymph Pressures In obstructive lymphedema, only a few lymphatic collectors remain patent. However, the tissue space is filled with fluid, and the remaining patent lymphatics are filled with lymph. The recorded pressures during rest range from 5 to 45 mm Hg, depending on the remaining contractility force of the damaged lymphatic musculature.12,13 During calf muscle contractions, pressures are generally low, ranging from 10 to 25 mm Hg; tiptoeing in some cases generates end pressures of more than 200 mm Hg. Although the end pressure can be high, flow is absent, because the proximal vessels are obstructed (Fig. 25-6). Massaging the foot or tapping the tissues containing lymphatics has no effect on lymph pressures. However, heating the foot significantly increases the pressure, amplitude, and frequency of lymphatic contractions.8,11

Lymph Flow Because most collectors are partially or totally obliterated in obstructive lymphedema, only a small amount of spontaneous flow is in patent vessel segments at different levels of the limb.12,13 In most cases, a correlation of pressure and flow shows ineffective contractions of lymphangions (see Fig. 25-6).

K23346_Neligan_25_Hydromechanics_r2_dc_0327-0048.indd 334

5/26/2015 9:46:37 AM

335

Chapter 25  Hydromechanics of Intercellular Fluid and Lymph

A

TISSUE FLUID PRESSURE: NORMAL LEG

mm Hg

4

2 0 Patient supine: no movement

Patient supine: movement

Patient supine: no movement

B

1 min

LYMPHEDEMA STAGE II

mm Hg

4

2

0 Patient supine: no movement

Patient supine: movement

Patient supine: no movement

Patient supine: after massage

FIG. 25-7  Tissue fluid pressures in normal and lymphedematous calf subcutaneous tissue in a horizontal position. A, In a normal leg, the pressure is approximately 0 mm Hg and is not affected by muscular contractions. B, In a lymphedematous limb, the pressure is approximately 2 mm Hg, with minor fluctuations during calf muscle contractions. Tissue fluid pressure, even in advanced stages of lymphedema, is low because of expansion of the interstitial space of the subcutis.

Subcutaneous Tissue Fluid Pressures Normal Tissue Under normal conditions, tissue fluid (intercellular fluid) pressure in the lower limb subcutaneous tissue measured at rest ranges between 23 and 11 mm Hg (Fig. 25-7). This is similar to findings from animal studies. Active calf movements may slightly decrease the pressure through emptying of the interstitial space; however, these differences are of no clinical importance.12

Lymphedema Tissue fluid pressure was measured under the skin in lymphedematous calf subcutis in a horizontal position12,13 (Fig. 25-8). Without movements, tissue fluid pressure ranged from 21.5 to 110 mm Hg (mean 2.5 6 3.0 mm Hg SD), and in controls, it ranged from 21.8 to 3.0 mm Hg (mean 0.8 6 1.2 mm Hg SD). No statistically significant differences were observed between them, although in some advanced cases pressures were slightly higher than the mean value. No significant changes in

K23346_Neligan_25_Hydromechanics_r2_dc_0327-0048.indd 335

5/26/2015 9:46:37 AM

336

Part IV  Diagnosis of Lymphedema

A

B

MIDCALF

ANKLE

10

8

mm Hg

6

4

2

0 Level 1 Patient supine: no movement

Level 1 Patient supine: movement

Level 1 Patient supine: no movement

Level 2 Level 2 Level 2 Patient supine: Patient supine: Patient supine: movement movement movement

FIG. 25-8  Tissue fluid pressures in lymphedema stage IV. A, Recorded at the level of the midcalf. B, Recorded at the level of the ankle. Minor differences were noted, depending on the level of measurement. Muscular contractions had no effect.

pressure occurred based on the change from the horizontal to upright position. The mechanism of low pressure could be explained by high skin compliance in the early stages of edema formation, leading to expansion of the subcutaneous space through absorption of excess capillary filtrate not drained by lymphatics. The protein-accumulating matrix osmotically absorbed water, expanded, and stretched the skin. A large subcutaneous space was created.

Subcutaneous Tissue Fluid Flow Normal Tissue In normal subcutaneous tissue, no flow is detectable (with contemporary means) at rest or during walking or massage.12

Minimum Tissue Fluid Pressure to Start Flow A water venous pressure manometer was attached to a needle placed in the subcutis. Fluid contained in the manometer tubing began to flow at pressures above 30 mm Hg (30 6 6 mm Hg).

Lymphedematous Tissue During muscular contractions in lymphedematous tissue, excess tissue fluid moves radially from the site of applied force but not unidirectionally toward the root of the extremity unless the distal portion of the limb is immediately bandaged.

K23346_Neligan_25_Hydromechanics_r2_dc_0327-0048.indd 336

5/26/2015 9:46:37 AM

Chapter 25  Hydromechanics of Intercellular Fluid and Lymph

337

Effects of Various Compression Techniques Compression therapy requires awareness of which tissue structures should be compressed to move edema fluid. When lymphatic collectors are occluded, lymph nodes fibrose. Edema tissue fluid accumulates in the spontaneously formed tissue spaces. Limb compression cannot push fluid back into the occluded/obliterated collectors. Compression forces move fluid along tissue spaces2,14 (Fig. 25-9). They act on skin, subcutaneous tissue, veins, lymphatics, and solid elements of tissue, such as fibers and ground matrix (Fig. 25-10). Deformation of these elements by external force

Pathways of tissue fluid flow to hypogastrium

Tissue fluid enters femoral canal

FIG. 25-9  A lymphedematous limb with obstructed lymphatics, spontaneously formed tissue spaces, and pathways of edema fluid flow to the hypogastrium and femoral canal. Such limbs need intensive compression therapy to evacuate excess tissue fluid.

Tissue spontaneous channels Obliterated lymphatics

Artery

Lymphatic

Pressure on vein by muscles Pressure on lymphatics

Vein

Pressure on skin and subcutis Bandage IPC

x?

Pressure on veins

Pressure on tissue fluid Capillary filtration

FIG. 25-10  Anatomic structures and lymph and tissue fluid locations in the foot and calf, with forces acting on tissues during manual compression, bandaging or intermittent pneumatic compression (IPC). Plasma filtration processes takes place in the exchange capillary vessels (blue arrows). Filtered fluid accumulates in the intercellular space (long red arrow pointing to the green question mark). Lymphatic collectors are occluded (red X). Compression forces act on skin, subcutaneous tissue, veins, lymphatics and the solid elements of tissue as fibers and ground matrix (red horizontal arrows). Deformation of these elements transfers force to the tissue fluid (long red oblique arrow). A large part of externally applied force dissipates on the solid elements before it increases tissue fluid pressure and moves it. Compression force should be high enough to move fluid along tissue spaces (green arrow) but should not occlude arteries and veins. Calf tissue deformation during compression procedures is indicated by the dashed lines.

K23346_Neligan_25_Hydromechanics_r2_dc_0327-0048.indd 337

5/26/2015 9:46:37 AM

338

Part IV  Diagnosis of Lymphedema

50

20

0

100 50

40

mm Hg

0

100 60

50 0 100 50

80

0

50 100 0 Patient supine: no movement

Patient supine: movement

FIG. 25-11  The effect of bandaging on intralymphatic pressures in a patient with obstructive stage II or III lymphedema of the lower limb.

K23346_Neligan_25_Hydromechanics_r2_dc_0327-0048.indd 338

5/26/2015 9:46:38 AM

Chapter 25  Hydromechanics of Intercellular Fluid and Lymph

339

transfers force to the mobile tissue fluid. A large portion of applied force is dissipated by the solid elements before it increases tissue fluid pressure and generates flow. Applied force should be high enough to move fluid without occluding arteries and veins. Effective tissue fluid pressures should exceed 30 mm Hg. This can be achieved by applying compression pressures of 50 to 60 mm Hg in early stages of lymphedema, and pressures of up to 125 mm Hg in advanced stages with stiff skin.

Manual Massage of the Foot The foot is the part of the lower limb where most tissue fluid and lymph are produced under normal conditions and accumulate in lymph flow obstruction. Manual massage of the foot in patients with early stages of lymphedema may propel lymph. However, discontinuation of massage is followed by an immediate cessation of lymph flow.

Bandaging of the Foot A gradual increase in bandage pressure raises lymph pressure to 50 mm Hg, with a series of lowamplitude pulses.13 Contractions of the calf muscle increase pressure to even higher levels (above 100 mm Hg). The most effective bandage pressures are 40 to 50 mm Hg. Higher pressures have no additional effect (Fig. 25-11). In the patient recorded in Fig. 25-11, a sphygmomanometer cuff had been placed around the foot and was wrapped by an elastic bandage. The intracuff pressures were gradually increased, and end pressures were simultaneously measured in a cannulated calf lymphatic vessel. Raising cuff pressures caused an increase of mean lymphatic pressure to 40 to 50 mm Hg. Intralymphatic pressures did not increase further, even with external pressures of 80 to 100 mm Hg. Contractions of calf muscles increased spontaneous pressures to 110 mm Hg at a cuff pressure of 40 mm Hg. Higher cuff pressures during calf muscle contractions had no additional effect on lymphatic pressure. The lymphangion’s contracting force reached peak values.

Manual Massage of the Calf Manual massage of lymphedematous calf soft tissues dissipates force radially. It may increase the tissue fluid pressure to higher than 100 mm Hg (Fig. 25-12). However, cessation leads to an immediate drop of pressure to 0 mm Hg. In advanced stages of lymphedema, high skin rigidity limits the force transferred to the subcutis, and thus limits lower pressures. To determine whether the manual force is transferred to proximal regions of the limb, we measured pressures at various distances from the massaging hand. Compression at a distance of 3 cm from the sensor did not raise pressure. This could be explained by low skin compliance and low hydraulic conductivity of the subcutis in lymphedema. Flow occurred only during hand pressure on the calf. Immediately after the hand was removed from the skin, flow stopped, with a backflow to the previously compressed area (Fig. 25-13).

K23346_Neligan_25_Hydromechanics_r2_dc_0327-0048.indd 339

5/26/2015 9:46:38 AM

340

Part IV  Diagnosis of Lymphedema

80

60 mm Hg

FIG. 25-12  Tissue fluid pressure during manual massage at three calf levels: above the ankle, in the midcalf, and under the knee. Sensors were inserted into the subcutaneous tissue. Massage was performed over the sensors. Each peak corresponded to one hand compression. Pressures reached different levels, as high as 100 mm Hg. The therapist was not informed of the level of pressure.

40

20

0 Massage above ankle Massage midcalf Massage under knee

FIG. 25-13  Plethysmographic recording of circumference changes with a strain gauge placed proximal and distal to hand compression (15 mm 5 5 mm limb circumference increase 5 10 to 15 ml tissue fluid flow). During compression, fluid moved proximally. Flow stopped after the hand was removed from the skin. Massage proximal to the strain gauge caused backflow (arrows).

Millimeters

15

10

5

0

Massage at ankle

Massage above ankle

Massage below knee

Massage above knee

Bandaging of the Calf One-layer bandaging with semistretch bandages raised tissue fluid pressures to 40 to 60 mm Hg. However, the generated pressures varied at different calf levels, depending on the mass of the soft tissue. They were usually lower under the knee and very low in the thigh (Fig. 25-14). Subsequent wrappings generated lower pressure, most likely because of the decrease of edema fluid volume. Pressure dropped slowly but significantly over time. This could partly be prevented by two-layer wrappings, because there is less elasticity with two layers than one. The level of tissue fluid pres-

K23346_Neligan_25_Hydromechanics_r2_dc_0327-0048.indd 340

5/26/2015 9:46:38 AM

341

Chapter 25  Hydromechanics of Intercellular Fluid and Lymph

70 60

FIG. 25-14  Tissue fluid pressure in the subcutaneous tissue during bandaging (short stretch) at three calf levels: above the ankle, in the midcalf, and under the knee. The sensors were inserted under the skin. Pressure varied at different levels and was lowest below the knee. Foot movements generated pressure waves of 20 mm Hg (mid part of the yellow curve). The pressure decreased at all levels after 5 minutes.

50

mm Hg

40 30 20 10 0 210 Massage midcalf Massage under knee Massage above ankle

Sites of tissue fluid pressure and limb circumference recording

FIG. 25-15  Schematic presentation of the lower limb in a pneumatic sleeve with eight chambers, each 9 cm long. Tissue fluid pressure was measured at six points (red dots). The lowest point in the calf was at the level of chamber 3. In the thigh, pressures were measured at chamber levels 6, 7, and 8. The lines encircling the calf and thigh indicate the site of strain gauge placement for continuous measuring of circumference changes during compression.

3

4

5

6

7

8

Pressure sensor

1

2

3

4

5

6

Sleeve chamber numbers

7

8

Strain gauge

sure during bandaging did not correlate with bandage markers showing the bandage-skin interface pressure.

Intermittent Pneumatic Compression Pneumatic compression has become a generally used modality.15-17 We use a multichamber pneumatic compression device that provides sequential inflation with gradient inflation pressure and an inflation time sufficient for translocation of tissue fluid to the proximal region. The distal chambers are not inflated, preventing fluid backflow and venous stasis in the superficial limb system. The device is composed of eight segments, each 9 cm long. Inflation pressures are regulated from 50 to 125 mm Hg. Gradient pressures decrease proximally by 20%. The inflation time of each chamber is 50 seconds, with a total inflation time of 400 seconds. The distal chambers are not deflated. The deflation time of all chambers is 50 seconds at the end of each cycle (Fig. 25-15).

K23346_Neligan_25_Hydromechanics_r2_dc_0327-0048.indd 341

5/26/2015 9:46:38 AM

342

Part IV  Diagnosis of Lymphedema

The Foot and Lower Part of the Leg In stage II and early stage III lymphedema, the effect of pneumatic compression was similar to that of manual massage. In some cases it evoked spontaneous lymphatic pulse waves. In stage IV, cannulation of lymphatics was usually unsuccessful. In some cases lymphatic contractility was absent.

The Calf and Thigh Tissue fluid pressures were lower than those in the chambers during the first inflation of each sleeve chamber in normal subjects and those with lymphedema18 (Figs. 25-16 and 25-17). The high gradient across the skin was most likely caused by skin rigidity (fibrosis) and dispersion of the applied force to the proximal noncompressed regions. Unexpectedly, little tissue fluid pressure was transmitted from the compressed to the noncompressed proximal segments for a distance of 9 cm (width of the chamber) in normal and lymphedematous limbs. For example, inflation of chambers 1 and 2 in the calf did not increase pressure at level 3. In lymphedematous limbs the popliteal and upper thigh tissue fluid pressures reached lower levels than in other limb regions. These two regions contain loose connective tissue, are usually less swollen, and accumulate fluid translocated during sequential massage.

Effects of Various Sequential Inflation Times on Tissue Fluid Pressure and Flow Tissue Fluid Pressures During Pneumatic Compression for 5 Seconds/Chamber Inflation Time Using a sequential inflation time of 5 seconds, and based on the recorded pressure curves, we were not able to discriminate the tissue fluid head pressures that were generated by consecutive chambers, regardless of whether the inflation pressure was 50, 80, or 120 mm Hg18 (Fig. 25-18).

120 100

mm Hg

80 Massage below knee Massage above ankle Massage midcalf

60 40 20 0

Calf 3 4 5 6 7 8

FIG. 25-16  Tissue fluid pressure recordings in normal calf subcutaneous tissue during pneumatic compression of 120 mm Hg, 50 seconds/chamber. Pressures were recorded at level 3, above the ankle; at chamber level 4, at the midcalf; and at level 5, below the knee. Tissue flow pressures during the first inflation of chambers at levels 3, 4, and 5 were lower than those in the chambers (black dots). Inflation of chamber 3 increased tissue fluid pressure to 40 mm Hg at the corresponding level. This increased to 80 mm Hg during inflation of consecutive chambers. Inflation of chamber 4 increased tissue fluid pressure to 60 mm Hg at this level. This increased to 100 mm Hg during inflation of consecutive chambers. Tissue pressure below the knee (level 5) rose to 100 mm Hg.

K23346_Neligan_25_Hydromechanics_r2_dc_0327-0048.indd 342

5/26/2015 9:46:38 AM

Chapter 25  Hydromechanics of Intercellular Fluid and Lymph

343

160 140 120

mm Hg

100 Massage midthigh Massage above knee Massage upper thigh

80 60 40 20 0 Thigh 3  4  5  6  7  8

FIG. 25-17  Tissue fluid pressure in the normal thigh subcutaneous tissue during pneumatic compression of 120 mm Hg for 50 seconds per chamber. Pressures were recorded in the chamber at level 6, above the knee; in the chamber at level 7, at the midthigh; and in the chamber at level 8, the upper thigh. Inflation of chamber 6 produced pressure of 110 mm Hg at level 6. Inflation of chamber 7 produced tissue fluid pressure of 110 mm Hg at level 7. The yellow curve shows a pressure of 25 mm Hg at level 8, close to the groin, usually with less edema and low flow resistance.

120 100

mm Hg

80 60 40 20

40 sec

0 220

50

80

120 mm Hg

FIG. 25-18  Tissue fluid pressures during 5 seconds of chamber pneumatic compression. Inflation of chambers 3, 4, and 5 generated tissue fluid head pressures under each chamber as low as 10 to 20 mm Hg, although the chamber pressures were much higher. Inflation pressures were 50, 80, and 120 mm Hg. Inflation of chambers 3, 4, and 5 generated tissue fluid head pressures as low as 10 to 20 mm Hg, although the chamber pressures were much higher. Inflation of the whole sleeve to 50 mm Hg, 80 mm Hg, and 120 mm Hg created final tissue fluid pressures of 25 to 40 mm Hg, 35 to 70 mm Hg, and 70 to 100 mm Hg, respectively. Inflation of the whole sleeve for 40 seconds did not generate and maintain tissue fluid pressure to a level as high as that in the sleeve. Moreover, a deflation period of 5 sec was not long enough to decrease pressure to 0 mm Hg. It remained at 20 to 30 mm Hg.

K23346_Neligan_25_Hydromechanics_r2_dc_0327-0048.indd 343

5/26/2015 9:46:39 AM

344

Part IV  Diagnosis of Lymphedema

Tissue Fluid Pressures During Pneumatic Compression for 20 Seconds/Chamber Inflation Time Using a sequential inflation time of 20 seconds, we were able to roughly discriminate between the tissue head pressures18 (Fig. 25-19).These pressures ranged from 10 to 20 mm Hg with chamber pressures of 50, 80, and 120 mm Hg. Tissue Fluid Pressures During Pneumatic Compression for 50 Seconds/Chamber Inflation Time In contrast to inflation times of 5 and 20 seconds, an inflation time of 50 seconds generated easily distinguishable tissue fluid head pressures at each level1 (Fig. 25-20). The mean head pressures under chamber 3 were 35, 45, and 60 mm Hg with inflation pressures of 50, 80, and 120 mm Hg, respectively. The respective pressures in the thigh were evidently lower, reaching 20 mm Hg in the groin. The head pressures at various limb levels depended on the shape of the limb dictated by the mass of soft tissues. They were high at the midcalf, lower below and above the knee, and higher at the midthigh, becoming low in the groin region. Inflation of the whole sleeve to 50 mm Hg created a pressure gradient between the calf and groin of 50 to 60 mm Hg to 20 mm Hg, respectively; inflation of the whole sleeve to 80 mm Hg created a pressure gradient of 85 to 30 mm Hg, respectively; and inflation of the whole sleeve to 120 mm Hg generated a pressure gradient of 130 to 40 mm Hg, respectively18 (Fig. 25-21). During deflation, the tissue fluid pressure dropped to 0 mm Hg, facilitating venous blood and tissue flow inflow into the distal limb segments to be compressed during the consecutive cycle.

120 120 mm Hg

100

mm Hg

80 60 40 20 0 220

160 sec

345678

FIG. 25-19  Tissue fluid pressures during 20 seconds per chamber of pneumatic compression. Inflation of the whole sleeve to 50 mm Hg generated tissue fluid pressures ranging from 50 mm Hg in the calf to 10 mm Hg in the groin. Inflation of the whole sleeve to 80 mm Hg generated tissue pressures ranging from 80 mm Hg in the calf to 20 mm Hg in the groin. Pressure generated by inflated chambers at various levels could not be discriminated because of short inflation times. Inflation of the whole sleeve to 120 mm Hg generated a tissue pressure of 40 mm Hg in the upper thigh. On deflation, tissue fluid pressure decreased to 20 to 30 mm Hg and did not reach the preinflation level of 0 to 5 mm Hg.

K23346_Neligan_25_Hydromechanics_r2_dc_0327-0048.indd 344

5/26/2015 9:46:39 AM

Chapter 25  Hydromechanics of Intercellular Fluid and Lymph

345

100 80

mm Hg

60 40 20 0

50 12345678

120

80

220

FIG. 25-20  Tissue fluid pressure recording in a lymphedematous calf subcutaneous tissue at the level of sleeve chambers 3 (orange curve), 4 (red curve), and 5 (blue curve). Inflation pressures were 50, 80, and 120 mm Hg. Each chamber was inflated for 50 seconds. Distal chambers were not deflated. Tissue fluid pressures during the first inflation were lower than those in the chambers. Inflation of chamber 3 increased tissue fluid pressure at level 3 to 30 mm Hg but not at level 4. Inflation of chamber 4 produced tissue fluid pressure at level 4 to 40 mm Hg and at level 5 to 12 mm Hg. The pressure curves had a similar shape during inflation to 80 and 120 mm Hg. At level 5 (third ascending line) close to the knee with soft popliteal tissue, the tissue fluid pressures were much lower than in chamber 5. This could be explained by low resistance to flow in the popliteal fossa. However, the most important observation was the lack of transmission of pressure from the compressed level to the next proximal level.

50 sec 3

130

4

5

6

7

8

120 110 100 90 mm Hg

80 70 60 50

120 80 60

40 30 20 10 0

FIG. 25-21  Data from 20 measurements of tissue fluid pressure during intermittent pneumatic compression, 50 seconds per chamber. The first inflation of consecutive chambers 3 to 8 generated head pressures of 50, 80, and 120 mm Hg (mean 6 SD, n 5 15). Tissue fluid pressures were lower than those in the chambers, especially below and above the knee and under chamber 8 in the groin.

K23346_Neligan_25_Hydromechanics_r2_dc_0327-0048.indd 345

5/26/2015 9:46:39 AM

346

Part IV  Diagnosis of Lymphedema

30 25 20 Millimeters

15 10 5 0 25 220 3 Ankle

4

5 Leg

6

7

8

Knee

FIG. 25-22  Circumference changes in the lower limb during pneumatic compression for 50 seconds per chamber. The volume can be determined from the increase in circumference. Flow was different at various levels of the limb, depending on the volume of soft tissues and contained fluid. It was highest in the midcalf and lower in the midthigh. Pressure and flow curves are similar in Figs. 25-21 and 25-22.

Tissue Fluid Flow During Pneumatic Compression for 50 Seconds/Chamber Inflation Time Flow was different at various levels of the limb depending on the volume of soft tissues and contained fluid. It was highest in the midcalf and lower in the midthigh (Fig. 25-22). This might be explained by the movement of accumulated fluid from the calf to the popliteal fossa and to the upper thigh where fluid dissipated to the neighboring regions.18

Conclusion This study showed that in obstructive lymphedema, the bulk of the stagnant tissue fluid is located deep in the subcutaneous tissue but not in the subepidermal lymphatic plexus. The resting tissue fluid pressures were slightly above 0 mm Hg. The minimum transmural (through skin and subcutis) tissue fluid pressures necessary for propelling tissue fluid in the subcutaneous tissue were found to be 30 6 15 mm Hg. The tissue fluid pressures generated by routine manual massage ranged from 40 to above 100 mm Hg, depending on the level of the leg massaged, although apparently the same compression force was applied. Variations in tissue fluid pressures during massage could be explained by differences in calf skin and subcutaneous tissue tonicity and thickness at various levels. Our studies showed that pressures generated by manual compression were high enough to overcome tissue hydraulic resistance and propel tissue fluid. However, on release of the hand, pressures dropped to 0 mm Hg. The rapidly disappearing pressure gradient must have resulted in cessation of fluid flow and even its backflow. The obtained data indicate that manual compression–generated tissue fluid pressures could be effective in propelling fluid. However, to maintain the unidirectional proximal flow, compression should last longer than 15 seconds, and fluid return should be prevented by immediate and tight compression.

K23346_Neligan_25_Hydromechanics_r2_dc_0327-0048.indd 346

5/26/2015 9:46:39 AM

Chapter 25  Hydromechanics of Intercellular Fluid and Lymph

347

Pneumatic compression of tissues with lymph stasis is beside manual massage a commonly used therapeutic modality in limb lymphedema. A number of pneumatic devices have been constructed. We tried to determine how high of a compression pressure should be applied, and for how long, to the limb soft tissues to reach tissue fluid head pressure above 30 mm Hg, necessary to initiate proximal flow. We found that (1) the tissue fluid head pressures were lower than those in inflated chambers, (2) inflation times of 5 and 20 seconds was too short to generate tissue fluid head pressures above 30 mm Hg, even if the compression pressures were as high as 120 mm Hg, (3) the 50-second timing achieved head pressures higher than 30 mm Hg. However, they consistently remained lower than in the compression chamber, (4) tissue fluid head pressures differed at various levels of the limb, depending on the soft tissue mass, (5) deflation of the inflated whole sleeve for 5 and 20 seconds was followed by high end pressures, whereas deflation of 50 seconds brought about a pressure drop to 0 mm Hg, facilitating refilling with tissue fluid of the distal parts of the massaged limb. Our observations point to the necessity of applying high pressures and compression times longer than 50 seconds to generate effective tissue fluid pressures and to provide enough time to facilitate flow.

C linical P earls • Lymph flow in limb lymphatics depends entirely on spontaneous rhythmic contractions of a segment between two unidirectional valves (lymphangions). • In lymphedema, there is no lymph flow in the collecting lymphatic vessels. • In lymphedema, tissue fluid accumulates in tissue spaces. • To move stagnant tissue fluid, compression in necessary. • Force (pressure/area) applied by compression materials and devices must be high enough to move fluid. • The effective compression pressure should be above 60 mm Hg in early stages of lymphedema and 125 mm Hg or higher in advanced stages. • Timing of compression is crucial for fluid movement because of tissue poroelastic resistance. Longer times are more effective. • High forces applied to tissues by bandages and devices are safe, because they have vertical direction and do not produce shear stress that damages microstructure.

R EFERENCES 1. Olszewski WL, Jain P, Ambujam G, Zaleska M, et al. Topography of accumulation of stagnant lymph and tissue fluid in soft tissues of human lymphedematous lower limbs. Lymphat Res Biol 7:239-245, 2009. 2. Zaleska M, Olszewski WL, Cakala M, et al. Intermittent pneumatic compression enhances formation of edema tissue fluid channels in lymphedema of lower limbs. Lymphat Res Biol. 2015 Mar 6. [Epub ahead of print]

K23346_Neligan_25_Hydromechanics_r2_dc_0327-0048.indd 347

5/26/2015 9:46:39 AM

348

Part IV  Diagnosis of Lymphedema

3. Olszewski WL, Jain P, Ambujam G, Zaleska M, et al. Tissue fluid pressure and flow during pneumatic compression in lymphedema of lower limbs. Lymphat Res Biol 9:77-83, 2011. 4. Engeset A, Harger B, Nesheim A, et al. Studies on human peripheral lymph. I. Sampling method. Lymphology 6:1-5, 1973. 5. Olszewski WL. Collection and physiological measurements of lymph and interstitial fluid in man. Lymphology 10:137-145, 1977. 6. Olszewski WL, Engeset A. Intrinsic contractility of leg lymphatics in man. Preliminary communication. Lymphology 12:81-84, 1979. 7. Olszewski WL, Engeset A. Lymphatic contractions. N Engl J Med 300:316, 1979. 8. Olszewski WL, Engeset A. Intrinsic contractility of prenodal lymph vessels and lymph flow in human leg. Am J Physiol 239:H775-H783, 1980. 9. Armenio S, Cetta F, Tanzini G, et al. Spontaneous contractility in the human lymph vessels. Lymphology 14:173-178, 1981. 10. Sjöberg T, Norgren L, Steen S. Contractility of human leg lymphatics during exercise before and after indomethacin. Lymphology 22:186-193, 1989. 11. Olszewski WL. Lymph vessel contractility. In Olszewski WL, ed. Lymph stasis: pathophysiology, diagnosis and therapy. Boca Raton, FL: CRC Press, 1991. 12. Olszewski WL. Contractility patterns of normal and pathologically changed human lymphatics. Ann N Y Acad Sci 979:52-63, 2002. 13. Olszewski WL. Contractility patterns of human leg lymphatics in various stages of obstructive lymphedema. Ann N Y Acad Sci 1131:110-118, 2008. 14. Olszewski WL, Cwikla J, Zaleska M, et al. Pathways of lymph and tissue fluid flow during intermittent pneumatic massage of lower limbs with obstructive lymphedema. Lymphology 44:54-64, 2011. 15. Rockson SG. Accruing evidence for a beneficial role of pneumatic biocompression in lymphedema. Lymphat Res Biol 8:v, 2010. 16. Pilch U, Wozniewski M, Szuba A. Influence of compression cycle time and number of sleeve chambers on upper extremity lymphedema volume reduction during intermittent pneumatic compression. Lymphology 42:26-35, 2009 17. Mayrovitz HN. Interface pressures produced by two different types of lymphedema therapy devices. Phys Ther 87:1379-1388, 2007. 18. Zaleska M, Olszewski WL, Jain P, et al. Pressures and timing of intermittent pneumatic compression devices for efficient tissue fluid and lymph flow in limbs with lymphedema. Lymphat Res Biol 11:227232, 2013.

K23346_Neligan_25_Hydromechanics_r2_dc_0327-0048.indd 348

5/26/2015 9:46:39 AM

C hapter 26 Radionuclide Lymphoscintigraphy Byung-Boong Lee, James Laredo

K ey P oints • Radionuclide lymphoscintigraphy (LSG) is an essential, noninvasive test not only for the diagnosis of chronic lymphedema, but also for the follow-up assessment of functional changes in lymphatic dynamics. • LSG is easy to perform, repeatable with reliable results, and harmless to the lymphatic endothelial lining. • LSG has been the procedure of choice for the clinical evaluation of lymphatic functional status and provides clear images of lymph transport vessels and draining nodes.

Chronic lymphedema is no longer viewed as a simple condition of static swelling of an affected limb or region after blockage of the lymph transporting and collecting systems. Lymphedema is now considered a progressive condition involving the lymphatic system and the entire skin and soft tissue integument in which a chronic degenerative and inflammatory process occurs, resulting in fibrotic change. This condition involves tissues beyond the lymphatics and lymph nodes, characterized by recurring episodes of dermatolymphoadenitis.1-4 Such a steadily progressing condition that affects the entire soft tissue envelope will eventually result in a disabling and distressing condition associated with numerous complications, including bacterial and fungal infections, dermatolipofibrosis with chronic inflammation, immunodeficiency and a wasting phenomenon, and malignancy (for example, Kaposi sarcoma and lymphangiosarcoma)5-8 (Fig. 26-1). Therefore chronic lymphedema is no longer considered a simple phenomenon of fluid accumulation but the beginning of a complex systemic disease. This mandates correct diagnosis at the earliest possible stage for more effective treatment. Radionuclide lymphoscintigraphy (LSG) fulfills a critical role as a precise method of evaluation and repeated assessment of patients with lymphedema for advanced treatment and care.9-12 One of the main advantages of LSG is that it is readily available at most centers, compared with some of the newer techniques, such as MRI and indocyanine green lymphangiography. Although these latter techniques in some ways provide

Rad

349

K23346_Neligan_26_Radionuclide_r4_0349-0364.indd 349

5/26/2015 9:50:41 AM

350

Part IV  Diagnosis of Lymphedema

FIG. 26-1  These three clinical cases represent advanced stage (clinical stage III) lymphedema with increasing morbidity by dermatolipofibrosis with chronic inflammation and persistent bacterial and fungal infections. When lymphedema reaches this end stage, it often becomes a life-threatening condition, with recurrent sepsis.

even better information than LSG, their relatively restricted availability and evolving role mean that LSG is still the benchmark procedure. Many conditions cause swelling (edema) that mimics lymphedema. True lymphedema as the cause of limb swelling is less common than one would think. Most cases of limb swelling are caused by local (cellulitis and arthritis), regional (deep vein thrombosis and chronic venous disease), and systemic disorders (congestive cardiac failure, renal failure, hypoalbuminemia, and protein-losing nephropathy). Moreover, lymphedema is not necessarily an isolated condition and can coexist with other medical conditions.13-16 In this context, LSG is essential not only to sort out the differential diagnosis, but also to evaluate the various conditions that can be associated with lymphedema (either primary or secondary), although a careful history and physical examination are often sufficient to make the diagnosis of chronic lymphedema.1,4 LSG is performed particularly for the assessment of primary lymphedema, because this congenital condition is the clinical manifestation of a truncular lymphatic malformation (LM) (Fig. 26-2). Together with extratruncular LM lesions, known as lymphangiomas, truncular LM malformations can occur in combination with many other vascular malformations (for example, a venous malformation).17-20 Thus a detailed evaluation of primary lymphedema should be performed, including the use of LSG. Other vascular imaging studies should also be done throughout all potential areas of involvement, as well as the contralateral normal limb or body part for comparison, because various

K23346_Neligan_26_Radionuclide_r4_0349-0364.indd 350

5/26/2015 9:50:41 AM

351

Chapter 26  Radionuclide Lymphoscintigraphy

B 30 min

A

1 hr

Uptake (%):

4.65

9.97

5.87

6.25

6.63

7.26

Uptake (%):

2.13

7.60

2.27

5.51

1.76

4.73

Clearance (%):

16.7

C



2 hr

20.6

21.7

22.7

32.0

33.0

D

Anterior

Anterior 70%

Posterior

Posterior 70%

FIG. 26-2  A, Left lower extremity with primary lymphedema caused by a combination of truncular LM and venous malformation, resulting in a discrepancy in leg length. Primary lymphedema was confirmed with extensive dermal backflow/lymph stasis shown in LSG. B, An infiltrating extratruncular venous malformation was also confirmed along the left lower leg, seen here in whole-body blood pool scintigraphy. C. It was also confirmed by the MRI. D, Such a combination of various vascular malformations warrants precise assessment of the lymphatic system with LSG, not only for the initial diagnosis, but also for the follow-up assessment of treatment and its natural course and progression.

abnormal conditions of lymphatic leakage, lymphangiectasia, chylous ascites, and chylothorax are infrequently involved.1,4,21,22 The basic initial diagnostic procedures for both primary and secondary lymphedema, including volumetric measurements and symptom evaluation, should be performed to make the earliest possible diagnosis in patients at risk.23-26 An assessment of the clinical condition, including the clinical staging of lymphedema, requires the use of LSG, along with some other basic diagnostic tests. This is especially the case for primary lymphedema.27

K23349_Neligan_26_Radionuclide_r4_0349-0364.indd 351

6/4/2015 1:11:40 PM

352

Part IV  Diagnosis of Lymphedema

Definition LSG is an imaging technique developed for the assessment of lymphatic function. It is a radionuclide technique that uses radioisotope-tagged or radioisotope-labeled pharmaceutical particles as a tracer, which are injected intradermally or subcutaneously.28-31 LSG is a functional study to complement the anatomic information on the lymphatic system gleaned from other studies, such as phlebography or venography. It offers not only an anatomic study of the subaponeurotic lymphatic vessels but also a functional assessment. One of the criticisms of LSG is that the anatomic definition of the images is very poor, and this is true in the context of surgical planning. Both MRI (see Chapter 28) and ICG lymphangiography (see Chapter 27) provide much better information on detailed anatomy. However, neither of these studies is as widely available as LSG. With LSG, an evaluation can be done in both qualitative and quantitative assays, and the quantitative measurements in particular can give functional imaging of the lymphatic transport capacity30-33 (Fig. 26-3). LSG, which was first introduced in 1953, has become increasingly popular because of the benign nature of the technique. LSG is a minimally invasive test, easily performed and safe. This method has largely replaced the more invasive and technically demanding technique of lymphangiography, although in the future it will likely be replaced by other higher definition studies. Nevertheless, LSG is now considered an essential test not only to confirm clinically suspected lymphedema, but also to assess the progress and response to treatment. LSG has replaced the classic role of oil contrast–based lymphangiography.28,29 Before LSG became easily available for the management of lymphedema, the classic oil contrast– based lymphangiography, which was established by Kinmonth,34 was the only tool to visualize the lymphatics. For more than 40 years, it was the procedure of choice for definitive delineation of the lymphatic system. However, three major issues associated with lymphography limited its widespread use: (1) difficulty in the cannulation of large lymphatic draining collectors through tiny skin lymphatics; (2) the risk of damage to the lymphatic endothelial lining by the iodinated oil contrast; and (3) the risk of oil (fat) embolism.35 Because it is a difficult technique with potential side effects, lymphangiography has fallen out of favor and has largely been abandoned as a diagnostic tool. It is seldom used today.26 Although LSG is considered a “functional” imaging modality, it delivers comprehensive and consistent images, visualizing various structural and functional changes in lymphatic flow dynamics. LSG produces dynamic images of lymphatic transport and the peripheral and central lymphatic structures and function. Its transport index score36 allows semiquantification of peripheral lymphatic radiotracer transport. Delayed imaging shows lymph node uptake, albeit without the detailed structural information seen with conventional lymphography.

K23346_Neligan_26_Radionuclide_r4_0349-0364.indd 352

5/26/2015 9:50:42 AM

353

Chapter 26  Radionuclide Lymphoscintigraphy

A

Number of lymph nodes

Normal

Moderately decreased

Severely decreased

None

Normal

Hypoplastic

None

Collateral

Normal

Mild

Moderate

Severe

B

Lymphatic vessel

C

Dermal backflow

FIG. 26-3  Criteria for the qualitative analysis of LSG. A, The number of lymph nodes (normal, moderately decreased, severely decreased, or none visible). B, The condition of the lymphatic vessels (normal, hypoplastic, none, or collateral). C, The extent of dermal backflow (normal, mild, moderate, or severe).

K23346_Neligan_26_Radionuclide_r4_0349-0364.indd 353

5/26/2015 9:50:42 AM

354

Part IV  Diagnosis of Lymphedema

TABLE 26-1  Guideline Criteria for the Laboratory Staging System (Grades I-IV)* Lymph Node Uptake

Dermal Backflow

Collateral Lymphatics

Main Lymphatics

Clearance of Radioisotope From Injection Site

Grade I

Decreased (6)

None (2)

Good visualization (1)

Decreased visualization (6)

Decreased lymphatic transport (6)

Grade II

Decreased to none (2)

Visualization (1)

Decreased visualization (6)

Poor to no visualization (6)

More decreased (6)

IIA: Extent of dermal backflow does not exceed one half of each limb IIB: Extent of dermal backflow exceeds one half of each limb

Grade III

No uptake (2)

Visualization (1)

Poor visualization (2)

No visualization (2)

No clearance (2)

Grade IV

None (2)

Poor to no visualization (2)

No visualization (2)

No visualization (2)

No clearance (2)

*Minimum of two or more LSG findings for laboratory staging.

Periodic LSG findings provide proper clinical and laboratory staging, which is essential for proper clinical management37,38 (Table 26-1). LSG also has special merit in assessing the efficacy of medicines, surgery, and physical means to facilitate lymph movement or reduce lymph formation before and after treatment. Images of truncal lymph transport and draining nodes can be routinely obtained for follow-up studies to document functional changes in lymphatic dynamics (Fig. 26-4). However, LSG has never been fully appreciated for its ability to evaluate the lymphatic system as an independent test or adjunct to the patient’s history and physical examination. This is partly related to clinician unfamiliarity with LSG or a bias against LSG based on the old concept of chronic lymphedema. Thus the appropriate use of LSG requires a proper understanding of the new concept of chronic lymphedema.1,4 LSG has now been well proved to be a safe, noninvasive, easy to perform method of assessing the lymphatic system. Furthermore, it is harmless to the lymphatic endothelial lining. In addition, technetium-99m (99mTc) has a short half-life (6 hours) and is nearly completely decayed within 24 hours. The diagnostic value of LSG is further enhanced when combined with MRI and/or duplex ultrasonography as one of three essential noninvasive and minimally invasive tests for the evaluation of the lymphatic system. This combination of tests improves lymphatic functional assessment, allows the evaluation of the involvement of the arterial and venous systems, and provides a framework for subsequent therapy.1,4

K23346_Neligan_26_Radionuclide_r4_0349-0364.indd 354

5/26/2015 9:50:42 AM

355

Chapter 26  Radionuclide Lymphoscintigraphy

A

B

C

D

FIG. 26-4  A, Right lower extremity in grade I LSG stage of lymphedema with mildly decreased lymphatic transport function. Decreased lymph node uptake, localized/minimum lymphatic retention, and dermal backflow are seen on a 2-hour image, although the main vessel still appears normal. On the contrary, the left lower extremity shows no evidence of lymphedema—grade 0 LSG stage with normal lymph node uptake, no dermal backflow, and well-visualized main lymphatic vessels. B, Right lower extremity in grade II LSG stage of lymphedema with partial lymphatic obstruction. Image shows much decreased lymph node uptake, a significant amount of dermal backflow exceeding one half of the extremity (grade IIB), scanty collateral lymphatics, and almost no visualization of the main vessel. C, Right lower extremity in grade III LSG stage of lymphedema with severe lymphatic obstruction. Image shows no lymph node uptake, nonvisualization of the main lymphatic vessel, and a significant amount of dermal backflow. D, Left lower extremity in grade IV LSG stage of lymphedema with nearly nonfunctioning lymphatics. Image shows no lymph node uptake, nonvisualization of the main lymphatic vessel, and no dermal backflow.

In addition to its primary role in assessing chronic lymphedema, recent advances in the understanding of phlebolymphedema as a combination of chronic venous insufficiency and chronic lymphatic insufficiency mandate a new role for LSG in the assessment of its lymphatic component, along with MRI and duplex ultrasonography.39,40 Simultaneous assessment of the venous and lymphatic systems is essential for the proper identification of the delicate relationship between these two inseparable and mutually interdependent systems. Therefore a proper understanding of the unique relationship between these three tests is necessary, especially for primary lymphedema and phlebolymphedema management. Duplex ultrasonography should be the first test performed, even before LSG, in all forms of primary lymphedema to differentiate between lymphatic and venous etiologic factors. The ultrasonic features of lymphedema include volumetric changes, a change in the thickness of the dermis and subcutaneous layer, and structural changes, such as a hyperechogenic dermis and hypoechogenic subcutaneous layer. The suprafascial and subfascial thicknesses of the edematous tissue in high resolution are very useful measurements that allow the periodic assessment of the response to

K23346_Neligan_26_Radionuclide_r4_0349-0364.indd 355

5/26/2015 9:50:42 AM

356

Part IV  Diagnosis of Lymphedema

therapy, monitor a patient’s progress, and determine the prognosis.41-44 Ultrasonography has the distinct advantage of being noninvasive and easy to perform. However, it is very user dependent and demands a significant skill level on the part of the ultrasonographer. The features of MRI include circumferential edema with a typical honeycomb pattern, in addition to increased subcutaneous tissue volume with marked thickening of the dermis. MRI is also helpful to identify lymph nodes and enlarged lymphatic trunks and to differentiate the various causes of lymphatic obstruction in secondary lymphedema. The anatomic information from MRI provides a substantial complement to the functional assessment provided by LSG.45,46 A more detailed description of MR lymphangiography, including the most recent developments in this technology, is found in Chapter 28. Recently LSG and CT scans have been combined to improve diagnostic accuracy by retrieving functional (scintigraphy) and anatomic (CT) data together.47,48 The limb volume computation is accurate with this three-dimensional reconstruction of the limb by the volume rendering technique49 to characterize excess fluid limited to the skin and subcutaneous tissues.

Technical Aspects The imaging quality of LSG depends on the selection of an appropriate radiolabeled macromolecule or colloidal material, because particles greater than 10 nm in diameter are transported only by the lymphatics. 99mTc antimony colloid has been recognized as the most ideal small size colloid (3 to 30 nm) among the many different radiopharmaceutical particles used as a tracer. Unfortunately, it is no longer available in the United States because of the withdrawal of Food and Drug Administration approval. Therefore the next best available agent is 99mTc sulfur colloid (10 to 1000 nm), which is filtered to remove the larger particles, thereby creating a nearly uniform particle size (10 to 50 nm).28,29,50,51 However, 99mTc albumin colloid is another 99mTc-labeled colloid with a good reputation, because the albumin microcolloid has a reproducible colloid size distribution (95% is less than 80 nm), and there is ease of labeling. Its rapid clearance from the injection site makes it suitable for quantitative studies, and the injections are painless.28,29,51 Other available noncolloidal tracers include 99mTc human serum albumin, 99mTc dextran, and 99mTc human immunoglobulin. A dual mechanism clears the noncolloidal tracers from the injection site, with resorption into the capillaries and transport through the lymphatics, affecting diagnostic specificity in patients with lymphatic leakage. Therefore the noncolloidal tracers require different criteria for interpretation than those for colloidal tracers. However, 99mTc dextran is sufficient to diagnose ordinary lymphedema with no lymphatic leakage, whereas 99mTc sulfur colloid is a better tracer when the surgeon is looking for leakage sites. Furthermore, it takes more time with 99mTc dextran than with 99mTc sulfur colloid for dynamic acquisition and transport from the injection site. 99mTc phytate, which is another agent often used for scintigraphy, may have some advantages in sentinel LSG.4,28,29,50

K23346_Neligan_26_Radionuclide_r4_0349-0364.indd 356

5/26/2015 9:50:42 AM

Chapter 26  Radionuclide Lymphoscintigraphy

357

The injection technique will differ depending on the goal of the study and radiotracer used. The subcutaneous route is better than the intradermal route when looking for lymphatic leakage. On the other hand, a deeper injection may be preferable when looking for metastatic lymph nodes, which mostly involve deep lymph nodes. However, for routine studies of superficial lymphatics of the extremity, both subcutaneous and intradermal injections can be used with good outcomes.28,29 The radiopharmaceutical particles (for example, 99mTc dextran or 99mTc sulfur colloid) are first injected subcutaneously into the webs between the first, second, and third toes (or fingers), two sites per limb, and 37 MBq (1 mCi) per site. After the injection, patients complete a standardized exercise routine (for example, walk for more than 30 minutes) to facilitate the clearance. Generally whole-body and spot imaging are acquired with a gamma camera in a series starting 60 minutes after the injection.28,29,50,51 The routine imaging modality of LSG for lymphedema is based on a whole-body scan plus spot imaging. Although the information and findings from the whole-body scan are sufficient for a clinical diagnosis and assessment, spot imaging can give more detailed information. In addition to the whole-body scan and spot imaging, dynamic LSG is another excellent option for monitoring the time-activity curve of inguinal nodes. However, the protocol for LSG has not been standardized regarding the various radiotracers and radioactivity doses, different injection volumes, intracutaneous versus subcutaneous injection site, epifascial or subfascial injection, number of injections, different protocols of passive and active physical activity, varying imaging times, and static and/or dynamic acquisition techniques.28,29,52 In 2009 and 2013, the International Union of Phlebology recommended that LSG performed with a subcutaneous injection of 99mTc-labeled human serum albumin or 99mTc-labeled sulfur colloid into the first and second webspace of the toes or fingers was the test of choice to confirm or exclude lymphedema as the cause of chronic limb swelling.1,4 Movement of the colloid from the injection site, transition time to the knee, groin, or axilla, absence or presence of major lymphatic collectors, number and size of vessels and nodes, presence of collaterals and reflux, and symmetrical activity with the opposite side are recorded and used for interpretation (see Table 26-1). Semiquantitative assessment has been reported, and most recently the technique of quantitative assessment of transit time from the foot to the knee has also been validated1,4 (see Figs. 26-3 and 26-4).

Interpretation Criteria and Guidelines LSG shows the superficial lymph transport system—main, larger lymph vessels and nodes—as the basic architecture of the peripheral lymphatic system only. It does not show the deep lymph vessels carrying lymph from the nodes back to the blood circulation. LSG identifies abnormal lymphatic transport after lymphedema is fully established, and it shows slow or absent lymph flow and reflux (dermal backflow) areas. LSG also shows tracer uptake abnormalities in the lymph nodes, which detect abnormalities of the lymphatic system in the extremities quite accurately, regardless of the cause.28,29

K23346_Neligan_26_Radionuclide_r4_0349-0364.indd 357

5/26/2015 9:50:42 AM

358

Part IV  Diagnosis of Lymphedema

A

LT

LT Posterior RT

RT Anterior

B

FIG. 26-5  A, Normal clearance with normal main lymphatic vessels, but multiple collateral vessels on both lower extremities might suggest insufficient function of the main channel system alone to warrant the collaterals for the compensation, compatible with the clinical status. B, The clinical condition is in the early stage of lymphedema combined with lipedema, as shown, at the borderline of stage I and stage 0.

The first and most important finding to be verified is whether there is increased accumulation of radiotracer in the lymphatic system and surrounding soft tissue (for example, dermal backflow). Its positive findings indicate not only the existence of lymphedema but also the severity (for example, delayed clearance) and range or extension of the disease. For lower limb lymphedema, the appearance and uptake of the inguinal nodes are directly proportional to the severity of the condition. If the inguinal nodes are not seen on interval follow-up scanning, this often indicates a more advanced stage of the disease. Abnormal lymph nodes or an abnormal pattern of lymphatic drainage through the collaterals often indicate lymphatic dysfunction, especially in the primary lymphedema group, although they have limited significance as nonspecific signs28,29 (Fig. 26-5). Generally, the main criteria of scintigraphic diagnosis depend on whether the lymphatic channels are clearly visible and radiotracer is accumulated in the soft tissue and/or lymphatic vessels. There are many abnormal patterns of LSG. Normal LSG in a normal limb reveals the tracer at the injection site, but to a lesser degree than in the lymphedematous limb. Radiotracer is neither accumulated along the normal lymphatic drainage route in the soft tissue nor in the lymphatic vessels (Fig. 26-6).

K23346_Neligan_26_Radionuclide_r4_0349-0364.indd 358

5/26/2015 9:50:43 AM

Chapter 26  Radionuclide Lymphoscintigraphy

LT

LT Posterior RT

359

RT Anterior

3-hour delay

6-hour delay LT

LT Posterior RT

RT Anterior

15-minute delay

FIG. 26-6  Normal radiotracer (99mTc-filtered sulfur colloid) uptake in normal-appearing lymphatic channels is seen throughout the bilateral lower extremities. Bilateral groin and iliac chain lymph nodes are seen on 15-minute, 3-hour, and 6-hour delayed images. Radiotracer uptake in the liver and thoracic duct is seen at the 6-hour delay. No abnormal pooling of radiotracer and dermal backflow is seen in either leg.

K23346_Neligan_26_Radionuclide_r4_0349-0364.indd 359

5/26/2015 9:50:43 AM

360

Part IV  Diagnosis of Lymphedema

The test reveals various conditions of tracer clearance, including the presence of dermal backflow and asymmetry alteration in inguinoaxillary nodes (sensitivity 89%, specificity 96%).33 However, the accumulation of radioactivity in the soft tissues or lymphatic vessels is the most ominous and common pattern seen as a diagnostic criterion of lymphedema, in addition to the findings of abnormal lymph node images or poor or no groin nodal uptake in the diseased extremity.28,29,50,51 Although these findings are not directly related to the cause of the lymphedema, the presence of more of these findings indicates the severity of the disease.

Primary Lymphedema In addition to a clinical evaluation, LSG is now the most essential modality used in the diagnosis of primary lymphedema. LSG is extremely useful to identify the specific lymphatic abnormality and to visualize the lymphatic network. LSG can easily be repeated with minimal risk. Data and images obtained from the study identify lymphatic (dys)function, which is based on the visualization of the lymphatics, lymph nodes, dermal backflow, and semiquantitative data on radiotracer (lymph) transport.1,4 However, in primary lymphedema, there is poor definition of the lymphatic routes. There is also a delayed appearance of tracer in the regional lymph nodes and possible tracer dermal backflow in hypoplasia, whereas in aplasia there are no lymphatic routes and lymph nodes are not displayed. With quantification, a more precise uptake can be measured in the groin. This method needs a normal standard and protocol that includes “normal values of uptake” in the groin and “clearance values of the injection site.”28,29 Another method of investigating lymph transport is the use of a transport index, in which an index below 10 is normal. The index combines visual assessment of the following five criteria: the temporal and spatial distribution of the radionuclide, appearance time of the lymph nodes, and graded visualization of lymph nodes and vessels.53

Secondary Lymphedema Secondary lymphedema has various causes. The underlying abnormality is the obstruction or obliteration of lymph flow from an acquired source. In developed countries the most common cause is cancer treatment of the regional axillary, inguinal, or retroperitoneal lymph nodes, which are excised, irradiated, or otherwise destroyed along with surrounding lymphatic vessels, and occasionally the staging of breast cancer, melanoma, or gynecologic malignancies. Therefore the general finding is one of decreased or absent transport combined with various grades of dermal backflow. Sometimes collateral circulation, lymphocele, and lymphangioectases can also be seen (Fig. 26-7). Quantification is also useful, especially in comparison with the contralateral limb, to determine if a preexisting lymphatic impairment is already present. This is often the case in so-called postinfectious secondary lymphedema, which not infrequently proves to be a primary impairment.13,53-55

K23346_Neligan_26_Radionuclide_r4_0349-0364.indd 360

5/26/2015 9:50:43 AM

Chapter 26  Radionuclide Lymphoscintigraphy

A

LT

LT Posterior RT

361

RT Anterior

B

15-minute delay

FIG. 26-7  A, Lymphoscintigraphy with 15-minute delay of 2.0 mCi of 99mTc sulfur colloid given intradermally. Severely tortuous main lymphatic vessels with the collaterals along the right lower extremity in a 56-year-old morbidly obese woman whose body weight was 280 pounds. B, Swelling in both legs rapidly became worse after recurrent bouts of cellulitis for the past few years and reached clinical stage III. Findings from her first LSG from 10 years ago were normal, which suggests that the infection and her lipedema were crucial precipitating factors for such rapid progression of the lymphedema.

Recommendation In the IUA-ISVI’s “Consensus for Diagnosis Guideline of Chronic Lymphedema of the Limbs,” the expert panel recommended LSG, from a morphofunctional viewpoint, for the pretreatment assessment of the lymphatic system.26 The panel also recommended LSG for follow-up assessment of therapy and natural disease progression for comparison with baseline values at the start. The strength of that recommendation is 1 (strong), and the quality of that evidence is A (high).56,57 Although the absolute majority of the panel gave a 1 (strong) and an A (high) recommendation to LSG as a noninvasive test for the basic evaluation of lymphedema, the current quality of LSG remains controversial because of poor image resolution. LSG is unable to detect edema in the lymphedematous limb and infrequently misidentifies dermal backflow. It is also known to yield false-positive findings (for example, erysipelas) in up to 10% of studies. However, LSG still has a leading role as a functional test in the initial diagnosis of an enlarged limb, although it hardly displays the anatomic abnormalities of a lymph vessel and lymph node compared with the old oil contrast lymphography.

K23346_Neligan_26_Radionuclide_r4_0349-0364.indd 361

5/26/2015 9:50:44 AM

362

Part IV  Diagnosis of Lymphedema

C linical P earls • Radionuclide lymphoscintigraphy (LSG) fulfills a critical role as a precise method of pretreatment evaluation and repeated assessment of patients with lymphedema. • The advantages of LSG are that it is minimally invasive, easily performed, safe, and widely available. Thus LSG remains the procedure of choice. • LSG is a radionuclide imaging technique for the assessment of lymphatic function that uses radioisotope-tagged or radioisotope-labeled pharmaceutical particles as a tracer. • Duplex ultrasonography should be the first test performed, even before LSG, in all forms of primary lymphedema to differentiate between lymphatic and venous etiologic factors. • LSG is used mostly in patients with primary lymphedema, although other vascular imaging studies should also be conducted in all areas of potential involvement and the contralateral normal limb for comparison.

R EFERENCES 1. Lee BB, Andrade M, Bergan J, et al; International Union of Phlebology. Diagnosis and treatment of primary lymphedema. Consensus document of the International Union of Phlebology (IUP)-2009. Int Angiol 29:454-470, 2010. 2. Lee BB. Chronic lymphedema, no more step child to modern medicine! Eur J Lymphol 14:6-12, 2004. 3. Lee BB. Contemporary issues in management of chronic lymphedema: personal reflection on an experience with 1065 patients. Lymphology 38:28-31, 2005. 4. Lee BB, Andrade M, Antignani PL, et al. Diagnosis and treatment of primary lymphedema. Consensus document of the International Union of Phlebology (IUP)-2013. Int Angiol 32:541-574, 2013. 5. International Society of Lymphology. The diagnosis and treatment of peripheral lymphedema. 2009 Consensus document of the International Society of Lymphology. Lymphology 42:51-60, 2009. 6. International Lymphoedema Framework. Best Practice for the Management of Lymphoedema, ed 2, 2012. Available at www.lympho.org. 7. Lee BB. Classification and staging of lymphedema. In Tredbar LL, Morgan CL, Lee BB, eds. Lymphedema: Diagnosis and Treatment. London: Springer-Verlag, 2008. 8. Lee BB, Laredo J, Neville R, et al. Primary lymphedema and Klippel-Trenaunay syndrome. In Lee BB, Bergan J, Rockson S, eds. Lymphedema: A Concise Compendium of Theory and Practice. London: Springer-Verlag, 2011. 9. Lee BB, Kim DI, Whang JH, et al. Contemporary management of chronic lymphedema—personal experiences. Lymphology 35(Suppl):450-455, 2002. 10. Lee BB, Laredo J. Contemporary role of lymphoscintigraphy: we can no longer afford to ignore! [editorial] Phlebology 26:177-178, 2011. 11. Lee BB, Laredo J, Neville R. Combined clinical and laboratory (lymphoscintigraphic) staging. In Lee BB, Bergan J, Rockson S, eds. Lymphedema: A Concise Compendium of Theory and Practice. London: Springer-Verlag, 2011. 12. Michelini S, Campisi C, Gasbarro V, et al. National guidelines on lymphedema. Lymphology 55:238242, 2007. 13. Damstra RJ, van Steensel MA, Boomsma JH, et al. Erysipelas as a sign of subclinical primary lymphoedema: a prospective quantitative scintigraphic study of 40 patients with unilateral erysipelas of the leg. Br J Dermatol 158:1210-1215, 2008.

K23346_Neligan_26_Radionuclide_r4_0349-0364.indd 362

5/26/2015 9:50:44 AM

Chapter 26  Radionuclide Lymphoscintigraphy

363

14. Tiwari A, Cheng KS, Button M, et al. Differential diagnosis, investigation, and current treatment of lower limb lymphedema. Arch Surg 138:152-161, 2003. 15. Lee BB, Kim YW, Seo JM, et al. Current concepts in lymphatic malformation (LM). Vasc Endovasc Surg 39:67-81, 2005. 16. Szuba A, Razavi M, Rockson SG. Diagnosis and treatment of concomitant venous obstruction in patients with secondary lymphedema. J Vasc Intervent Radiol 13:799-803, 2002. 17. Lee BB, Laredo J. Classification: venous-lymphatic vascular malformation. In Allegra C, Antignani PL, Kalodiki E, eds. News in Phlebology. Turin, Italy: Edizioni Minerva Medica, 2013. 18. Lee BB, Villavicencio JL. Primary lymphoedema and lymphatic malformation: are they the two sides of the same coin? Eur J Vasc Endovasc Surg 39:646-653, 2010. 19. Lee BB, Laredo J, Seo JM, et al. Treatment of lymphatic malformations. In Mattassi R, Loose DA, Vaghi M, eds. Hemangiomas and Vascular Malformations. Milan: Springer-Verlag, 2009. 20. Lee BB. Lymphedema-angiodysplasia syndrome: a prodigal form of lymphatic malformation (LM). Phlebolymphology 47:324-332, 2005. 21. Pui MH, Yueh TC. Lymphoscintigraphy in chyluria, chyloperitoneum and chylothorax. J Nucl Med 39:1292-1296, 1998. 22. Cambria RA, Gloviczki P, Naessens JM, et al. Noninvasive evaluation of the lymphatic system with lymphoscintigraphy: a prospective, semiquantitative analysis in 386 extremities. J Vasc Surg 18:773782, 1993. 23. Bernas M, Witte M, Witte C, et al. Limb volume measurements in lymphedema: issues and standards. Lymphology 29(Suppl):199-202, 1996. 24. Sitzia J. Volume measurements in lymphoedema treatment: examination of formulae. Eur J Cancer Care (Engl) 4:11-16, 1995. 25. Casley-Smith JR. Measuring and representing peripheral oedema and its alterations. Lymphology 27:56-70, 1994. 26. Lee BB, Antignani PL, Baroncelli TA, et al. Iua-Isvi consensus for diagnosis of chronic lymphedema of the limbs. Int Angiol. 2014 Mar 19. [Epub ahead of print] 27. Gloviczki P, ed. Guidelines 6.2.0 on lymphoscintigraphy and lymphangiography. In Gloviczki P, ed. Handbook of Venous Disorders: Guidelines of the American Venous Forum, ed 3. London: Hodder Arnold, 2009. 28. Yuan Z, Chen L, Luo Q, et al. The role of radionuclide lymphoscintigraphy in extremity lymphedema. Ann Nucl Med 20:341-344, 2006. 29. Peller PJ, Bender CE, Gloviczki P. Lymphoscintigraphy and lymphangiography. In Gloviczki P, ed. Handbook of Venous Disorders: Guidelines of the American Venous Forum, ed 3. London: Hodder Arnold, 2009. 30. Szuba A, Shin WS, Strauss HW, et al. The third circulation: radionuclide lymphoscintigraphy in the evaluation of lymphedema. J Nucl Med 44:43-57, 2003. 31. Weissleder H, Weissleder R. Lymphedema: evaluation of qualitative and quantitative lymphoscintigraphy in 238 patients. Radiology 167:729-735, 1988. 32. Baulieu F, Lorette G, Baulieu JL, et al. [Lymphoscintigraphic exploration in the limbs lymphatic disease] Presse Med 39:1292-1304, 2010. 33. Infante JR, Garcia L, Laguna P, et al. Lymphoscintigraphy for differential diagnosis of peripheral edema: diagnostic yield of different scintigraphic patterns. Rev Esp Med Nucl Imagen Mol 31:237-242, 2012. 34. Kinmonth JB. Lymphangiography in man; a method of outlining lymphatic trunks at operation. Clin Sci (Lond) 11:13-20, 1952. 35. Steckei RJ, Furumanski S, Dunham R, et al. Radionuclide perfusion lymphangiography. An experimental technique to complement the standard ethiodol lymphangiogram. Am J Roentgenol Radium Ther Nucl Med 124:600-609, 1975. 36. Baumeister RG, Siuda S, Bull U, et al. Evaluation of transport kinetics in lymphoscintigraphy: followup study in patients with transplanted lymphatic vessels. Eur J Nucl Med 10:349-352, 1985.

K23346_Neligan_26_Radionuclide_r4_0349-0364.indd 363

5/26/2015 9:50:44 AM

364

Part IV  Diagnosis of Lymphedema

37. Lee BB, Bergan JJ. New clinical and laboratory staging systems to improve management of chronic lymphedema. Lymphology 38:122-129, 2005. 38. Michelini S, Failla A, Moneta G, et al. Clinical staging of lymphedema and therapeutical implications. Lymphology 35:168-176, 2002. 39. Lee BB, Laredo J, Neville R, et al. Diagnosis and management of primary phlebolymphedema. In Lee BB, Bergan J, Rockson S, eds. Lymphedema: A Concise Compendium of Theory and Practice. London: Springer-Verlag, 2011. 40. Lee BB, Bergan J, Gloviczki P, et al; International Union of Phlebology (IUP). Diagnosis and treatment of venous malformations. Consensus document of the International Union of Phlebology (IUP)-2009. Int Angiol 28:434-451, 2009. 41. Lim CY, Seo HG, Kim K, et al. Measurement of lymphedema using ultrasonography with the compression method. Lymphology 44:72-81, 2011. 42. Cavezzi A. Duplex ultrasonography. In Lee BB, Bergan J, Rockson S, eds. Lymphedema: A Concise Compendium of Theory and Practice. London: Springer-Verlag, 2011. 43. Antignani PL, Benedetti-Valentini F, Aluigi L, et al; Italian Society for Vascular Investigation. Diagnosis of vascular diseases. Ultrasound investigation—guidelines. Int Angiol 31(5 Suppl 1):1-77, 2012. 44. Garra BS. Imaging and estimation of tissue elasticity by ultrasound. Ultrasound Q 23:255-258, 2007. 45. Lu Q, Xu JR, Liu NF. Chronic lower extremity lymphedema: a comparative study of high-resolution interstitial MR lymphangiography and heavily T2-weighted MRI. Eur J Radiol 73:365-373, 2010. 46. Werner GT, Rodiek SO. Value of nuclear magnetic resonance tomography in leg edema of unknown origin. Preliminary report. Z Lymphol 17:2-5, 1993. 47. Buck AK, Nekolla S, Ziegler S, et al. SPECT/CT. J Nucl Med 49:1305-1319, 2008. 48. Kotani K, Kawabe J, Higashiyama S, et al. Lymphoscintigraphy with single-photon emission computed tomography/computed tomography is useful for determining the site of chyle leakage after esophagectomy. Indian J Nucl Med 27:208-209, 2012. 49. Uhl JF. 3D multislice CT to demonstrate the effects of compression therapy. Int Angiol 29:411-415, 2010. 50. Hung JC, Wiseman GA, Wahner HW, et al. Filtered technetium-99m-sulfur colloid for lymphoscintigraphy. J Nucl Med 36:1895-1901, 1995. 51. Inoue Y, Otake T, Nishikawa J, et al. Lymphoscintigraphy using Tc 99m human serum albumin in chylothorax. Clin Nucl Med 22:60, 1997. 52. Bellini C, Boccardo F, Campisi C, et al. Lymphatic dysplasias in newborns and children: the role of lymphoscintigraphy. J Pediatr 152:587-589, 2008. 53. Kleinhans E, Baumeister RG, Hahn D, et al. Evaluation of transport kinetics in lymphoscintigraphy: follow-up study in patients with transplanted lymphatic vessels. Eur J Nucl Med 10:349-352, 1985. 54. Mariani G, Campisi C, Taddei G, et al. The current role of lymphoscintigraphy in the diagnostic evaluation of patients with peripheral lymphedema. Lymphology 31(Suppl):316-319, 1998. 55. Bellini C, Di Battista E, Boccardo F, et al. The role of lymphoscintigraphy in the diagnosis of lymphedema in Turner syndrome. Lymphology 42:123-129, 2009. 56. Guyatt GH, Gutterman D, Baumann MH, et al. Grading strength of recommendations and quality of evidence in clinical guidelines. Chest 129:174-181, 2006. 57. Guyatt GH, Oxman AD, Vist GE, et al; GRADE Working Group. GRADE: an emerging consensus on rating quality of evidence and strength of recommendations. BMJ 336:924-926, 2008.

K23346_Neligan_26_Radionuclide_r4_0349-0364.indd 364

5/26/2015 9:50:44 AM

C hapter 27 Indocyanine Green Lymphography Mitsunaga Narushima, Takumi Yamamoto, Isao Koshima

K ey P oints • Indocyanine green (ICG) lymphography, which can visualize superficial lymph flow in real time without radiation exposure, is a clinically useful evaluation method for lymphedema. • With progression of lymphedema, the ICG lymphographic pattern changes from the normal linear pattern to abnormal dermal backflow (DB) patterns (splash, stardust, and diffuse patterns).

Ind

• ICG velocity and lymph transportation capacity decrease as lymphedema progresses. • ICG lymphography is also useful for the preoperative assessment of lymphatic surgeries, because different ICG lymphographic findings represent different conditions of the lymphatic vessels; the more severe the DB pattern seen on ICG lymphography, the more sclerotic and smaller the lymphatic vessels are. • In dynamic ICG lymphography, one ICG injection is enough for pathophysiologic severity staging (DB stage), lymph pump function evaluation (ICG velocity), and preoperative assessment.

Several methods have been reported to visualize lymph flow and evaluate lymphedema, including MRI, CT, ultrasonography, and lymphoscintigraphy.1-4 Currently, lymphoscintigraphy is considered the benchmark for the evaluation of lymphedema. Lymphoscintigraphy can visualize deep lymphatic flow but its image is obscure, and it has a risk of radiation exposure.4,5 ICG was first reported in 2007 for the evaluation of lymphedema.6,7 ICG lymphography allows much clearer visualization of superficial lymph flow than lymphoscintigraphy. It is useful not only for lymphedema evaluation but also for the preoperative assessment of lymphatic surgeries.6-16 ICG lymphography is increasingly used in the clinical practice of lymphedema management.

365

K23346_Neligan_27_Indocyonine_r3_dc_0365-0378.indd 365

5/26/2015 9:56:17 AM

366

Part IV  Diagnosis of Lymphedema

Material for Indocyanine Green Lymphography As an optical tracer agent, ICG is a medically useful green dye that was approved by the FDA in 1956. ICG is not only a green dye marker but also a fluorescent substance. Near-infrared fluorescent light penetrates tissue more deeply than does visible light.6,7 ICG lymphography, which is used to visualize superficial lymph flow after injection of ICG, is performed with a near-infrared camera device that is equipped with a charge-coupled device camera as a detector with a 760 nm light-emitting diode and a filter-cutting light below 820 nm.6-12 The fluorescent images are digitized for real-time display by using a standard personal computer. Several near-infrared camera devices are available. Some manufacturers include Hamamatsu Photonics, Mizuho, Novadaq Technologies, Olympus (microscope), Carl Zeiss (microscope), and Leica (microscope).13,14 Because ICG lymphography is used to visualize lymph flow in real time, ICG lymphography allows static evaluation of lymph circulation as well as dynamic lymph movement. With this property, dynamic ICG lymphography, or dual-phase ICG lymphography, was developed for comprehensive assessment of lymphedema.17,18

Procedures of Dynamic Indocyanine Green Lymphography Dynamic ICG lymphography is performed as follows. After a patient remains still for 15 minutes, 0.05 to 0.2 ml of ICG (Diagnogreen 0.25%, Daiichi Pharmaceutical, Tokyo, Japan) is injected subcutaneously (at the second web space for extremity and genital lymphedema and at the glabella and below the nose for facial lymphedema).8-12 Immediately after ICG injection (transient phase), fluorescent images of lymphatic flow are obtained with an infrared camera system.17,18 The observation is continued until lymph pump function is measured. Patients remain still in the supine position during lymph pump function measurement. Then patients are allowed to move freely. Twelve to 18 hours after ICG injection, ICG movement usually reaches the plateau phase. In this phase, lymph circulation is evaluated based on ICG lymphographic findings, which allow pathophysiologic severity staging and preoperative guidance for lymphatic surgeries.8-12 When patients move their extremity rigorously, ICG can reach a plateau 2 hours after ICG injection.17,18 The plateau phase usually continues until 72 hours after ICG injection. Thus it is possible to evaluate abnormal lymph circulation between 2 and 72 hours after ICG injection.15,16

K23346_Neligan_27_Indocyonine_r3_dc_0365-0378.indd 366

5/26/2015 9:56:17 AM

Chapter 27  Indocyanine Green Lymphography

367

ICG injection 5 minutes Remain still Early transient phase 2-72 hours Move freely

IGC velocity (pump)

Late plateau phase Dermal backflow stage (circulation) Mapping (navigation)

FIG. 27-1  Dynamic ICG lymphography. At the early transient phase, lymph pump function is evaluated by ICG velocity. At the late plateau phase, pathophysiologic severity staging (dermal backflow stage) and lymphatic mapping are done based on ICG lymphographic patterns.

In dynamic ICG lymphography, lymphatic images are taken twice; at an early transient phase and at a late plateau phase.17,18 We usually inject ICG the day before lymphatic surgery; one ICG injection is enough for lymph pump function measurement, evaluation of abnormal lymph circulation, and preoperative mapping (Fig. 27-1).

Evaluation of the Lymphatics Lymph Vessels As ICG lymphographic patterns change according to the pathophysiologic changes of lymph flows, different ICG lymphographic findings indicate different conditions of the lymphatic vessels.9-16 As ICG lymphographic patterns change from linear to splash, stardust, and diffuse pattern, lymphatic vessels change to become more sclerotic with less lymph flow. Lymphatic vessels are almost intact in linear regions; on the other hand, they are very sclerotic with a pinhole-like lumen in diffuse regions9-14 (Fig. 27-2). According to a comparative study of ICG lymphographic findings and intraoperative conditions of 215 lymphatic vessels, the mean diameter of the lymphatic vessels was 0.45 mm in linear regions, 0.44 mm in splash and stardust regions, and 0.26 mm in diffuse regions.15 In planning lymphaticovenular anastomosis, diffuse regions should be avoided, because the detection rate of lymphatic vessels favorable for lymphaticovenular anastomosis is low.15,16

K23349_Neligan_27_Indocyonine_r4_cl_0365-0378.indd 367

5/28/2015 12:59:03 PM

368

Part IV  Diagnosis of Lymphedema

Linear Normal

Splash

Stardust Abnormal (dermal backflow)

Diffuse

Progression of Lymphedema

FIG. 27-2  ICG lymphographic findings and conditions of lymphatic vessels. With progression of lymphedema, ICG lymphography patients change from linear to splash, stardust, and finally to a diffuse pattern.

Lymph Transportation Capacity Because ICG lymphography visualizes lymph flow in real time, lymph transportation capacity can be directly evaluated by measuring ICG movement at an early transient phase. There are several ways to evaluate lymph pump function, such as transit time, lymphatic pressure, and ICG velocity.17-20 Among the various lymph pump function evaluations, ICG velocity is the most practical one. ICG velocity can be measured within 5 minutes, whereas others sometimes require more than 1 hour in patients with severe lymphedema. The distance between the injection point and farthest proximal point at which the dye can be observed is measured 5 minutes after ICG injection; ICG velocity is calculated by dividing the distance by time. When ICG reaches the axilla and groin within 5 minutes after dye injection in patients with extremity lymphedema, ICG velocity is calculated by dividing the distance between the injection point and axilla and groin by the time required. ICG velocity decreases with the progression of lymphedema and increases after successful interventions. Because ICG velocity is a quantitative evaluation, it is easy to evaluate lymph pump function before and after therapeutic interventions.

Lymph Circulation ICG lymphography can detect lymphatic channels and nodes located up to 2 cm deep to the skin surface, assuming there is no fascia, muscle, or bone between the lymph channels and skin. Thus ICG lymphography dynamically shows superficial lymphatic channels (collectors, precollectors, and capillary channels) as a white line (linear pattern) on a screen. ICG lymphography can show lymphatic channels, lymphatic valves, and lymphatic flow immediately after the injection of ICG.

K23346_Neligan_27_Indocyonine_r3_dc_0365-0378.indd 368

5/26/2015 9:56:17 AM

369

Chapter 27  Indocyanine Green Lymphography

The ICG lymphographic pattern changes from the normal linear pattern to abnormal dermal backflow (DB) patterns in obstructive lymphedema. With the progression of lymphedema, ICG lymphographic patterns change from the linear pattern to the splash, stardust, and finally to the diffuse pattern.8-12 Lymph flow obstruction leads to lymphatic hypertension, lymphangiectasis, lymphatic valve insufficiency, lymphosclerosis, and lymph backflow. ICG lymphography visualizes lymph backflow as the DB pattern. The splash pattern on ICG lymphography represents dilated superficial lymphatics, such as lymphatic precollectors and capillaries. Extravasation of lymph fluid occurs with the progression of lymphedema, which is represented as spots on ICG lymphography, the stardust pattern. Finally, the number of spots visualized on ICG lymphography increases to the point at which spots merge and cannot be distinguished from each other, the diffuse pattern. These DB patterns usually extend from proximal to distal regions in obstructive lymphedema, such as cancer-related lymphedema. The evaluation of abnormal lymph circulation with ICG lymphography allows pathophysiologic severity staging, which is a qualitative evaluation.

Dermal Backflow Stage for the Pathophysiologic Severity Staging System The DB stage, the pathophysiologic severity staging system, is determined based on ICG lymphographic findings (Table 27-1). There are four DB stages9-12,16: 1. Arm DB (ADB) (Fig. 27-3) 2. Leg DB (LDB)10 (Fig. 27-4) 3. Genital DB (GDB)12 (Fig. 27-5) 4. Facial DB (FDB) (Fig. 27-6)

TABLE 27-1  Dermal Backflow Pattern Staging Arm (ADB): Upper Extremity

Leg (LDB): Lower Extremity

Genital (GDB): Lower Abdominal and Genital Regions

Facial (FDB): Head and Neck Regions

0

No DB pattern seen

No DB pattern seen

No DB pattern seen

No DB pattern seen

I

Splash pattern seen around axilla

Splash pattern seen around groin

Splash pattern seen around groin and/or lower abdominal region

Splash pattern seen around neck

II

Stardust pattern limited proximally to olecranon

Stardust pattern limited proximally to superior border of patella

Stardust pattern around groin and/or lower abdominal region

Stardust pattern limited proximally to mandibular margin

III

Stardust pattern beyond olecranon

Stardust pattern extends distal to superior border of patella

Stardust pattern present in whole area between groin and genital regions

Stardust pattern extends proximal to mandibular margin

IV

Stardust pattern observed throughout limb

Stardust pattern observed throughout limb

Stardust pattern observed throughout groin and/or lower abdominal regions

Stardust pattern observed throughout head and neck region

V

Diffuse stardust pattern

Diffuse stardust pattern

Stage

Diffuse stardust pattern

ADB, Arm dermal backflow; LDB, leg dermal backflow; GDB, genital dermal backflow; FDB, facial dermal backflow.

K23346_Neligan_27_Indocyonine_r3_dc_0365-0378.indd 369

5/26/2015 9:56:17 AM

370

Part IV  Diagnosis of Lymphedema

ADB Stage 0

ADB Stage III

Splash



Stardust



1 1

Splash Stardust Diffuse

Diffuse



1

1 1 1

ADB Stage IV

ADB Stage I

Splash



Splash Stardust Diffuse

Stardust Diffuse

1

ADB Stage II



1 1 1

1

ADB Stage V

Splash Stardust Diffuse

Splash Stardust Diffuse

FIG. 27-3  Arm dermal backflow (ADB) stage.

K23346_Neligan_27_Indocyonine_r3_dc_0365-0378.indd 370

5/26/2015 9:56:18 AM

371

Chapter 27  Indocyanine Green Lymphography



LDB Stage III

LDB Stage 0

Splash



Stardust





Splash

Diffuse



1



11

Stardust Diffuse

111

LDB Stage IV

LDB Stage I

Splash Stardust Diffuse





Splash



1



Stardust Diffuse

1 1 1

1

LDB Stage V

LDB Stage II

Splash

Stardust Diffuse

Splash

Stardust Diffuse

FIG. 27-4  Leg dermal backflow (LDB) stage.

K23346_Neligan_27_Indocyonine_r3_dc_0365-0378.indd 371

5/26/2015 9:56:18 AM

372

Part IV  Diagnosis of Lymphedema

GDB Stage 0

GDB Stage III

Splash



Stardust



1 1

Splash Stardust Diffuse

Diffuse



1

1 1 1

1

GDB Stage IV

GDB Stage I

Splash



Stardust Diffuse

Splash Stardust Diffuse

1

GDB Stage II

Splash Stardust Diffuse

FIG. 27-5  Genital dermal backflow (GDB) stage.

K23346_Neligan_27_Indocyonine_r3_dc_0365-0378.indd 372

5/26/2015 9:56:19 AM

373

Chapter 27  Indocyanine Green Lymphography

FDB Stage 0

FDB Stage III

Splash



Stardust



1 1

Splash Stardust Diffuse

Diffuse



1

1 1 1

FDB Stage IV

FDB Stage I

Splash



Splash Stardust Diffuse

Stardust Diffuse

1

FDB Stage II



1 1 1

1

FDB Stage V

Splash Stardust Diffuse

Splash Stardust Diffuse

FIG. 27-6  Facial dermal backflow (FDB) stage.

K23346_Neligan_27_Indocyonine_r3_dc_0365-0378.indd 373

5/26/2015 9:56:19 AM

374

Part IV  Diagnosis of Lymphedema

Linear

Splash

Stardust

Diffuse

Stage 0

Stage I

Stage II-IV

Stage V

Irreversible

Reversible Progression of Lymphedema

FIG. 27-7  Clinical significance of DB pattern differentiation. The splash pattern is a reversible lymph circulatory change, whereas the stardust pattern is an irreversible lymph circulatory change.

According to a prospective cohort study by Akita et al21 in which 100 consecutive patients with gynecologic cancer were followed for 2 years with ICG lymphography, the splash pattern was seen as a reversible lymph circulation change, whereas the stardust pattern was an irreversible change (Fig. 27-7). After pelvic lymph node dissection, 31 of 100 patients showed the splash pattern (LDB stage I), of which 5 improved to the linear pattern (LDB stage 0), 17 remained in the splash pattern (LDB stage I), and 9 progressed to the stardust pattern (LDB stage II). After observation on ICG lymphography, the stardust pattern never improved to the splash or linear pattern, even with conservative treatment. Because most patients are asymptomatic in DB stage I, asymptomatic patients should be followed carefully with ICG lymphography after cancer treatment to detect subclinical lymphedema.10,21,22

Intraoperative Navigation for Lymphaticovenular Anastomosis ICG lymphography can be used intraoperatively for navigation during lymphatic surgery. As mentioned previously, conventional ICG lymphography with a handheld camera system or microscopic ICG lymphography with a near-infrared camera-integrated microscope guides the surgeon

K23349_Neligan_27_Indocyonine_r4_cl_0365-0378.indd 374

5/28/2015 12:59:03 PM

Chapter 27  Indocyanine Green Lymphography

375

to find lymphatic vessels suitable for lymphaticovenular anastomosis, to evaluate the patency or quality of lymphaticovenular anastomosis, and to harvest a vascularized lymph node flap.13,14,16,23-32 In severe DB regions, such as those with a diffuse pattern, lymphatic vessels are less likely to be enhanced intraoperatively. Thus intraoperative ICG lymphography is likely to help a lymphatic surgeon in mild lymphedema cases.13,14,27 When observed at a plateau phase, additional ICG injection is unnecessary or does not work for intraoperative navigation. When lymphatic vessels are not enhanced, the vessels are usually sclerotic, and additional ICG injection will not enhance them.13,14

Lymphedema Management With Indocyanine Green Lymphography When to start treatment and which treatments to perform are the most important issues to be solved in lymphedema management. ICG lymphography can be a useful evaluation method to guide decision-making in lymphedema management16,22 (Table 27-2). In DB stage 0 (no lymphedema), no treatment is indicated, because lymph circulation is intact. In DB stage I with a splash pattern (subclinical lymphedema), management is controversial. Although the splash pattern represents an abnormal lymph circulation pattern, it is a reversible change. Immediate therapeutic interventions are unnecessary and overtreatment should be avoided.10,16,22 Further studies are required to clarify the management of subclinical lymphedema: prophylactic treatment or careful follow-up with ICG lymphography. In DB stage II with the stardust pattern (early lymphedema), physiologic surgical treatments, such as LVA, are indicated, because there is an irreversible abnormal lymph circulation pattern that is refractory to conservative treatments. When treatment is indicated, conservative treatment should be initiated first not only as a definitive treatment, but also as a preoperative modality before lymphatic surgery. When lymphedema progresses despite conservative treatment, lymphatic surgery is indicated. For subclinical or early-stage lymphedema (DB stages I and II), lymphaticovenular anastomosis is the best treatment option because of its effectiveness and minimal invasiveness. Lymphaticovenular anastomosis can be performed under local anesthesia through a small incision.* For more advanced *References 10, 13, 14, 16, 22, 27.

TABLE 27-2  Dermal Backflow Stage and Lymphedema Management DB Stage

Clinical Phase

Management

Stage I

Subclinical

Follow-up or prophylaxis

Stage II

Early

Lymphaticovenular anastomosis

Stages III-V

Progressed

Lymphaticovenular anastomosis 1 Vascularized lymph node transfer 1 Liposuction

K23349_Neligan_27_Indocyonine_r4_cl_0365-0378.indd 375

5/28/2015 12:59:03 PM

376

Part IV  Diagnosis of Lymphedema

lymphedema (DB stages III through V), lymphaticovenular anastomosis can work, but further surgical treatments are sometimes required when lymphaticovenular anastomosis is insufficient to improve the lymphedema. In particular, for DB stage V with a diffuse pattern, vascularized lymph node transfer is indicated, because there are few lymphatic vessels appropriate for lymphaticovenular anastomosis. Debulking surgeries, such as resection and liposuction, are required for volume reduction in patients with massive fat deposition.

C linical P earls • ICG lymphography follows cancer survivors’ prognosis regarding lymphedema. • The splash pattern (DB stage I) is a reversible lymph circulatory change; some patients improve to DB stage 0 spontaneously. • The stardust pattern (DB stage II) is an irreversible lymph circulatory change; no patients improve to DB stages 0 or I even with conservative treatment.

R EFERENCES 1. Case TC, Witte CL, Witte MH, et al. Magnetic resonance imaging in human lymphedema: comparison with lymphangioscintigraphy. Magn Reson Imaging 10:549-558, 1992. 2. Gamba JL, Silverman PM, Ling D, et al. Primary lower extremity lymphedema: CT diagnosis. Radiology 149:218, 1983. 3. Doldi SB, Lattuada E, Zappa MA, et al. Ultrasonography of extremity lymphedema. Lymphology 25:129133, 1992. 4. Henze E, Schelbert HR, Collins JD, et al. Lymphoscintigraphy with Tc-99m-labeled dextran. J Nucl Med 23:923-929, 1982. 5. International Society of Lymphology. The diagnosis and treatment of peripheral lymphedema. 2009 Consensus Document of the International Society of Lymphology. Lymphology 42:51-60, 2009. 6. Ogata F, Azuma R, Kikuchi M, et al. Novel lymphography using indocyanine green dye for nearinfrared fluorescence labeling. Ann Plast Surg 58:652-655, 2007. 7. Unno N, Inuzuka K, Suzuki M, et al. Preliminary experience with a novel fluorescence lymphography using indocyanine green in patients with secondary lymphedema. J Vasc Surg 45:1016-1021, 2007. 8. Yamamoto T, Narushima M, Doi K, et al. Characteristic indocyanine green lymphography findings in lower extremity lymphedema: the generation of a novel lymphedema severity staging system using dermal backflow patterns. Plast Reconstr Surg 127:1979-1986, 2011. 9. Yamamoto T, Yamamoto N, Ogata F, et al. Indocyanine green-enhanced lymphography for upper extremity lymphedema: a novel severity staging system using dermal backflow patterns. Plast Reconstr Surg 128:941-947, 2011. 10. Yamamoto T, Matsuda N, Doi K, et al. The earliest finding of indocyanine green lymphography in asymptomatic limbs of lower extremity lymphedema patients secondary to cancer treatment: the mod­ ified dermal backflow stage and concept of subclinical lymphedema. Plast Reconstr Surg 128:314e321e, 2011. 11. Yamamoto T, Iida T, Matsuda N, et al. Indocyanine green (ICG)-enhanced lymphography for evaluation of facial lymphoedema. J Plast Reconstr Aesthet Surg 64:1541-1544, 2011.

K23349_Neligan_27_Indocyonine_r4_cl_0365-0378.indd 376

6/4/2015 1:17:52 PM

Chapter 27  Indocyanine Green Lymphography

377

12. Yamamoto T, Yamamoto N, Yoshimatsu H, et al. Indocyanine green lymphography for evaluation of genital lymphedema in secondary lower extremity lymphedema patients. J Vasc Surg: Venous Lymph Dis. 2013 July 18. [Epub ahead of print] 13. Yamamoto T, Yamamoto N, Azuma S, et al. Near-infrared illumination system-integrated microscope for supermicrosurgical lymphaticovenular anastomosis. Microsurgery 34:23-27, 2014. 14. Yamamoto T, Yamamoto N, Numahata T, et al. Navigation lymphatic supermicrosurgery for the treatment of cancer-related peripheral lymphedema. Vasc Endovasc Surg 48:139-143, 2014. 15. Yamamoto T, Narushima M, Koshima I. Lymphedema evaluation using ICG fluorescent lymphography. In Koshima I, ed. Clinical Textbook of Lymphedema. Osaka: Nagai Shoten, 2011. 16. Yamamoto T, Yamamoto N, Narushima M, et al. Lymphaticovenular anastomosis with guidance of ICG lymphography. J Jpn Coll Angiol 52:327-331, 2012. 17. Yamamoto T, Narushima M, Yoshimatsu H, et al. Indocyanine green velocity: lymph transportation capacity deterioration with progression of lymphedema. Ann Plast Surg 71:591-594, 2013. 18. Yamamoto T, Narushima M, Yoshimatsu H, et al. Dynamic indocyanine green (ICG) lymphography for breast cancer-related arm lymphedema. Ann Plast Surg 73:706-709, 2014. 19. Unno N, Nishiyama M, Suzuki M, et al. Quantitative lymph imaging for assessment of lymph function using indocyanine green fluorescence lymphography. Eur J Endovasc Surg 36:230-236, 2008. 20. Unno N, Nishiyama M, Suzuki M, et al. A novel method of measuring human lymphatic pumping using indocyanine green fluorescence lymphography. J Vasc Surg 52:946-952, 2010. 21. Akita S, Mitsukawa N, Rikihisa N, et al. Early diagnosis and risk factors for lymphedema following lymph node dissection for gynecologic cancer. Plast Reconstr Surg 131:283-290, 2013. 22. Yamamoto T, Koshima I. Subclinical lymphedema: understanding is the clue to decision making. Plast Reconstr Surg 132:472e-473e, 2013. 23. Yamamoto T, Yoshimatsu H, Narushima M, et al. Split intravascular stents for side-to-end lymphaticovenular anastomosis. Ann Plast Surg 71:538-540, 2013. 24. Yamamoto T, Narushima M, Kikuchi K, et al. Lambda-shaped anastomosis with intravascular stenting method for safe and effective lymphaticovenular anastomosis. Plast Reconstr Surg 127:1987-1992, 2011. 25. Yamamoto T, Yoshimatsu H, Narushima M, et al. A modified side-to-end lymphaticovenular anastomosis. Microsurgery 33:130-133, 2013. 26. Yamamoto T, Koshima I, Yoshimatsu H, et al. Simultaneous multi-site lymphaticovenular anastomoses for primary lower extremity and genital lymphoedema complicated with severe lymphorrhea. J Plast Reconstr Aesthet Surg 64:812-815, 2011. 27. Yamamoto T, Narushima M, Yoshimatsu H, et al. Minimally invasive lymphatic supermicrosurgery (MILS): indocyanine green lymphography-guided simultaneous multisite lymphaticovenular anastomoses via millimeter skin incisions. Ann Plast Surg 72:67-70, 2014. 28. Yamamoto T, Yoshimatsu H, Narushima M, et al. Sequential anastomosis for lymphatic supermicrosurgery: multiple lymphaticovenular anastomoses on 1 venule. Ann Plast Surg 73:46-49, 2014. 29. Yamamoto T, Yoshimatsu H, Yamamoto N, et al. Side-to-end lymphaticovenular anastomosis through temporary lymphatic expansion. PLoS ONE 8:e59523, 2013. 30. Yamamoto T, Koshima I. Colourful indocyanine green lymphography. J Plast Reconstr Aesthet Surg 67:432-433, 2014. 31. Yamamoto T, Kikuchi K, Yoshimatsu H, et al. Ladder-shaped lymphaticovenular anastomosis using multiple side-to-side lymphatic anastomoses for a leg lymphedema patient. Microsurgery 34:404-408, 2014. 32. Narushima M, Mihara M, Yamamoto Y, et al. The intravascular stenting method for treatment of extremity lymphedema with multiconfiguration lymphaticovenous anastomoses. Plast Reconstr Surg 125:935-943, 2010.

K23346_Neligan_27_Indocyonine_r3_dc_0365-0378.indd 377

5/26/2015 9:56:20 AM

K23346_Neligan_27_Indocyonine_r3_dc_0365-0378.indd 378

5/26/2015 9:56:20 AM

C hapter 28 Magnetic Resonance Lymphangiography Lee M. Mitsumori

K ey P oints • Three-dimensional heavily T2-weighted sequences are performed to assess the severity, extent, and distribution of lymphedema. • The number, size, and location of individual subdermal lymphatic channels and the areas of dermal backflow can be visualized with magnetic resonance lymphangiography (MRL). • MRL can be performed on 1.5 T or 3.0 T platforms with clinically available, high-resolution, three-dimensional volumetric sequences used for MR angiography. • MRL requires the intracutaneous injection of an extracellular gadolinium-based MR contrast agent. • Image postprocessing of three-dimensional volumetric MR datasets facilitates examination interpretation.

Ma

A number of microsurgical procedures for the long-term treatment of lymphedema have been developed, and the importance of individualizing the type of operative treatment based on the degree of lymphatic dysfunction and the state of the subcutaneous tissue is being recognized.1-5 One of the current challenges for lymphatic surgery is that there is no standardized method of imaging the structure and function of the lymphatic circulation to evaluate the quality and severity of lymphedema.6 Two lymphatic imaging methods that are clinically available are nuclear medicine lymphoscintigraphy and indocyanine green fluorescence lymphography (see Chapters 26 and 27). Although radionuclide lymphoscintigraphy has been considered the primary clinical imaging modality to diagnose lymphedema,7 the limited temporal and spatial resolution of the modality does not allow the identification and localization of individual lymphatic channels.8,9 Indocyanine green fluorescence lymphography is an imaging modality that is frequently used intraoperatively for microsurgical lymphatic procedures. Although indocyanine green fluorescence lymphography can provide a real-time map of the subdermal lymphatic channels, it has several disadvantages for the preoperative evaluation of lymphedema. These include a small field of view,

379

K23349_Neligan_28_Mag Reso_r3_dc_379-392.indd 379

5/27/2015 9:33:45 AM

380

Part IV  Diagnosis of Lymphedema

lack of spatial information, limited skin penetration depth,6 and the inability to characterize soft tissues. Thus improved lymphatic imaging techniques are needed to determine whether suitable lymphatic channels are present for reconstruction, to depict the location of suitable lymphatic channels for preoperative planning, and to delineate the status of the subcutaneous soft tissues to facilitate the selection of the optimal treatment approach. Magnetic resonance lymphangiography (MRL) is a noninvasive technique that can image the subdermal lymphatic circulation in patients with lymphedema. MRL examinations provide the anatomic coverage to image an entire extremity with a high-resolution three-dimensional dataset and have sufficient temporal and spatial resolution to depict individual lymphatic channels and areas of dermal backflow. Imaging capabilities that are needed for the preoperative planning of lymphatic microsurgery.4,9 In addition, other MR pulse sequences can be included in the examination to evaluate the status of the subcutaneous soft tissues.8 With current 1.5 T and 3.0 T MR platforms, imaging data are acquired as three-dimensional volumetric datasets, which is important to enable the use of widely available image postprocessing algorithms that facilitate examination interpretation and microsurgical treatment planning.9,10 The main difference between MRL and other conventional contrast-enhanced MR examinations is the route of contrast administration. In MRL, a small volume of an extracellular gadoliniumbased MR contrast agent is injected intracutaneously in the interdigital webspaces of the hand or foot to promote contrast uptake by the lymphatic circulation. The low molecular weight of the extracellular MR contrast agents allows the lymphatic circulation to absorb the contrast agent in the interstitial space.11 Because of the off-label route of administration, the safety of the intracutaneous administration of extracellular gadolinium-based MR contrast agents was initially evaluated with animal experiments.12 Since the first few human clinical studies,13,14 several studies have been published that support the safety of the intracutaneous administration of different extracellular gadolinium-based MR contrast agents for MRL (gadopentetate dimeglumine,9 gadoterate meglumine,13 gadoteridol,4,15 gadodiamide,14,16 and gadobenate dimeglumine17,18). Although the intracutaneous contrast injection has been described as well tolerated by patients and no complications were reported by these studies, patients do describe mild to moderate pain during the injection, and some have transient swelling of the dorsum of the extremity around the injection site.11,13,14,16 To reduce the pain of the injection, a local anesthetic can be mixed with the contrast agent before intracutaneous administration, and a small-gauge needle should be used.11

MRL Imaging An MRL examination consists of two main components: a T2-weighted sequence to depict the severity and distribution of lymphedema and a fat-suppressed three-dimensional spoiled gradient recalled–echo (3D-SPGR) sequence after the intracutaneous injection of MR contrast to image lymphatic channels. Because the intracutaneously administered contrast agent is also absorbed by the venous circulation,1,9,11 the same high-resolution 3D-SPGR sequence can be repeated after a separate intravenous injection of contrast to obtain an MR venogram.19 Having the delayed MR

K23349_Neligan_28_Mag Reso_r3_dc_379-392.indd 380

5/27/2015 9:33:45 AM

Chapter 28  Magnetic Resonance Lymphangiography

381

venogram to compare with the MRL can be helpful to differentiate enhancing lymphatic channels from contrast-containing veins. MRL can be performed at 1.5 T or 3.0 T.9,15-17 Patient positioning, coil placement, and scan orientation will depend on whether the examination is of an upper or lower extremity and whether unilateral or bilateral imaging is performed. For unilateral imaging of the upper extremity, the patient is positioned supine and head first in the scanner gantry. The arm that will be scanned is placed at the patient’s side, and the patient is positioned as far laterally as possible with the arm propped with a pad to the level of the magnet isocenter. Surface coils are then positioned to image the target arm from the midhand to the shoulder. At 1.5 T we use a 16-channel torso phased array surface coil, whereas at 3.0 T a digital system with imbedded table coil elements is used, which allows coverage of the entire extremity with automatic coil element selection for each individual scan location. Headfirst patient positioning is helpful to allow access to the patient’s hand for the intracutaneous contrast injection that is performed midway through the examination. For the lower extremities, MRL is performed either as a unilateral or bilateral examination, depending on the clinical request. Patients are placed supine and feet first on the scanner table to allow access to the patient’s feet from the far side of the gantry for the intracutaneous contrast injection. At 1.5 T a 16-channel torso phased array surface coil is used, and a two-station examination is performed.16 The surface coil is initially positioned to cover the lower legs from the midfoot to above the knees and then repositioned to cover the thighs from the knees to the groin. Alternatively, a dedicated peripheral vascular surface coil can be used to image the upper and lower legs simultaneously.14 At 3.0 T we use a digital system that allows automatic coil element selection for the desired scan range, and the study is performed as a three-station examination. Because inhomogeneous fat suppression can produce regions of high signal that can obscure lymphatic channels and areas of dermal backflow9 (Fig. 28-1), patient position with unilateral examinations is offset laterally to place the extremity of interest as close as possible to the magnet isocenter, which improves shimming and the uniformity of fat suppression. Two primary sequences are included in our MRL examination: (1) a heavily T2-weighted threedimensional turbo spin-echo with spectral fat suppression (spectral presaturation with inver­ sion recovery) to define the severity and extent of edema and (2) a dynamic T1-weighted threedimensional spoiled gradient–recalled echo (3D T1w GRE) with fat suppression before and after the intracutaneous contrast injection to visualize enhancing lymphatic channels and dermal backflow (Figs. 28-1 and 28-2). Example MR sequence parameters used at our institution are presented in Table 28-1. Currently we perform seven dynamic phase acquisitions at approximately 5-minute intervals (0, 5, 10, 15, 20, 25, and 30 minutes).17After these initial dynamic phases are acquired, the images are reviewed to determine if additional phases are needed to include the entire extent of lymphatic enhancement. After the MRL, we perform an MR venogram to help differentiate subdermal lymphatics from veins. For the MR venogram, a single phase of the 3D T1w GRE scan used for

K23349_Neligan_28_Mag Reso_r3_dc_379-392.indd 381

5/27/2015 9:33:45 AM

382

A

Part IV  Diagnosis of Lymphedema

B

C

D

E

DBF3

FIG. 28-1  This 60-year-old woman had a left mastectomy for breast cancer and presented with progressive left arm swelling. A and B, Coronal full-volume maximum intensity projection (MIP) and single-plane multiplanar reformation (MPR) reconstructed images of the left arm from the T2-weighted sequence demonstrate moderate epifascial edema in the lateral upper arm (arrow). C and D, Coronal full-volume MIP and single-plane MPR reconstructed images of the contrast-enhanced MRL depict areas of skin enhancement representing dermal backflow in the proximal, mid, and distal left arm (arrowheads). In C the scalloped areas of high signal on MRL (*) represent regions of inhomogeneous fat suppression. Full-volume MIP reconstructions are helpful to depict the extent and spatial distribution of the findings, whereas single-plane MPR images are used to define the location of a finding and exclude artifacts. Interactive examination review with image postprocessing software allows co-localization of a finding on different sequences and the creation of different viewing planes. E, Reconstructed axial MPR image from the MRL scan data made through the distal area of dermal backflow (arrowhead) seen on the full-volume MIP. The axial MPR confirms that this reflects skin enhancement (DBF3), which is located along the posterior medial surface of the arm. The patient was considered a candidate for lymphaticovenular bypass.

the MRL is repeated 120 to 180 seconds after an intravenous injection of MR contrast. The threedimensional scans of the upper or unilateral lower extremities are performed in the sagittal orientation for more efficient scanning. The three-dimensional scans of the bilateral lower extremities are performed in the coronal orientation for the larger lateral scan coverage needed to include both lower extremities in the same scan field of view. Currently at our institution, the average examination time for an upper or lower extremity MRL is between 1.5 and 2 hours. The same contrast injection protocol is used at both field strengths. The intracutaneous injection of MR contrast consists of a mixture of 10 ml of contrast (gadobenate dimeglumine) with 1 ml of 1% lidocaine and 1 ml of sodium bicarbonate.17 The contrast-anesthetic mixture is then drawn into a 5 ml syringe for a unilateral examination or a 10 ml syringe for a bilateral examination. The skin of the dorsal hand or foot is sterilely prepared, and 1 ml of the contrast-anesthetic mixture is injected intracutaneously into each of the four interdigital webspaces of the hand or foot with a

K23349_Neligan_28_Mag Reso_r3_dc_379-392.indd 382

5/27/2015 9:33:45 AM

Chapter 28  Magnetic Resonance Lymphangiography

383

TABLE 28-1  Example Sequence Parameters of MRL T2w 3D TSE

3D T1w GRE (1.5 T)

3D T1w GRE (3.0 T)

Sequence

3D Multishot TSE

3D T1-TFE

3D T1-FFE

Orientation

Sagittal

Sagittal

Sagittal

Profile order

Reverse linear

Low-high radial

Linear

Half-scan factor

0.8

Fat suppression

SPIR

SPIR

Dual-echo Dixon

Field of view

380 3 312 3 150 mm3

485 3 162 3 100 mm3

360 3 221 3 147 mm3

Voxel size

1.7 3 1.7 3 3.0 mm3

1.3 3 1.3 3 1.0 mm3

1.2 3 1.2 3 1.6 mm3

TR

2500 ms

7.2 ms

6.2 ms

TE

350 ms

3.3 ms

1.5 ms/2.8 ms

Flip angle

90 degrees

30 degrees

20 degrees

SENSE factor

2 in AP direction

None

None

Scan time

3:47 per station

1:27 per dynamic

1:25 per dynamic

0.85 3 0.85

These sequence parameters reflect typical values for the scanner platform used at our institution. 3D, Three-dimensional; FFE, fast field echo; SENSE, Sensitivity encoding; SPIR, spectral presaturation with inversion recovery; T2w, T2weighted; TE, echo time; TFE, turbo field echo; TR, repetition time; TSE, turbo spin-echo.

26-gauge needle (total 4 ml per extremity). After the intracutaneous contrast has been administered, the injection sites are massaged for 60 seconds to promote lymphatic uptake.7,16 The intravenous contrast administration for the MR venogram can be performed manually or with a power injector typically in an antecubital vein and is composed of 0.1 mmol/kg of contrast (gadobenate dimeglumine) injected at a rate of 1.0 ml/sec followed by a 20 ml saline flush at the same rate. Although several different gadolinium-based MR contrast agents have been used for MRL,4,9,13,16,17 we elected to use gadobenate because of the higher relaxivity, thermal stability, and potential for protein binding that this contrast agent provides.20,21

Examination Interpretation and Image Postprocessing The objectives of MRL are to define the severity and extent of lymphedema and to depict the number, course, and location of enhancing lymphatic channels and dermal backflow. For examination interpretation, we interactively review the source images on an independent workstation to allow the real-time creation and viewing of three-dimensional postprocessed images. MPR, MIP reconstructions, volume rendering, and the three-dimensional cursor are used interactively to characterize and colocalize a particular structure on different image reconstructions. To evaluate the presence and severity of peripheral lymphedema, multiplanar reformatted images of the three-dimensional heavily T2-weighted images are reviewed together with a full-volume MIP reconstruction of the scanned extremity18 (see Figs. 28-1 and 28-2, A and B). We describe the location and extent of the edema and qualitatively grade the severity of the edema as mild,

K23349_Neligan_28_Mag Reso_r3_dc_379-392.indd 383

5/27/2015 9:33:45 AM

384

Part IV  Diagnosis of Lymphedema

A C

D

B

FIG. 28-2  This 72-year-old woman had a left mastectomy with axillary lymph node dissection and radiotherapy. She presented with progressive left arm swelling. A, Coronal full-volume MIP reconstruction of the T2 sequence of the left arm reveals diffuse high signal intensity throughout the arm, reflecting severe lymphedema. B, Corresponding axial MPR reconstructed T2 image of the distal forearm shows the circumferential distribution and severity of the edema. C and D, Coronal full-volume MIP images of the left arm before and 30 minutes after intracutaneous contrast injection in the hand. The MRL shows multiple enhancing irregular lymphatic channels throughout the arm. The patient was considered a candidate for lymphaticovenular bypass.

moderate, or severe. Next we examine the dynamic contrast-enhanced three-dimensional MRL, in which lymphatic channels are typically seen as irregular, discontinuous, or twisted16,17 (see Figs. 28-1 and 28-2, C and D). Enhanced lymphatic channels may not be seen in normal limbs on MRL, which is thought to be the result of faster lymphatic transport in a healthy limb (Fig. 28-3).17 Full-volume MIPs are created for each dynamic phase to select the phase with the greatest degree of lymphatic channel enhancement. Subsequent image review is then performed on the selected phase with the greatest degree of lymphatic enhancement. Because small patient movements frequently occur during the 5-minute pauses between MRL scan phases, subtraction techniques19 are frequently distorted by misregistration artifacts and thus are not currently created. For reporting MRL examination findings, different general patterns of lymphatic drainage have been described17 and example cases are

K23349_Neligan_28_Mag Reso_r3_dc_379-392.indd 384

5/27/2015 9:33:45 AM

385

Chapter 28  Magnetic Resonance Lymphangiography

Before contrast

5 minutes

10 minutes

15 minutes

20 minutes

30 minutes

FIG. 28-3  This 35-year-old man had left lower extremity edema and was referred for lymphaticovenular bypass microsurgery. Fused two-station coronal full-volume MIP reconstructed images from the multiphase MRL acquired at different time points—before contrast administration and then at 5, 10, 15, 20, and 30 minutes after intracutaneous contrast injection in the left foot. Images show irregular radiating lymphatic channels extending from the left ankle to the left groin. No enhancing lymphatic channels were seen in the right leg. The degree of lymphatic enhancement can vary with time. In this patient, the greatest lymphatic enhancement was observed at 5 minutes. Note the peripheral scalloped areas of high signal along both thighs, reflecting regions of incomplete fat suppression. The patient was considered a candidate for lymphaticovenular bypass.

K23349_Neligan_28_Mag Reso_r3_dc_379-392.indd 385

5/27/2015 9:33:45 AM

386

Part IV  Diagnosis of Lymphedema

A

B

C

D

FIG. 28-4  Different MRL patterns of lymphatic drainage in lymphedematous limbs. A, Sagittal MIP of a left arm showing discontinuous, minimally enhancing lymphatic channels (arrowheads). B, Sagittal MIP of a left leg showing dilated lymphatic channels with few branches (arrowheads). C, Coronal oblique MIP of a left leg showing radiating, clustered enhancing lymphatic channels that assemble at the medial knee and continue proximally into the thigh (arrowheads). D, Coronal MIP of a left arm showing numerous reticular skin lymphatics and areas of dermal backflow (arrowheads). Incomplete fat suppression appears as scalloped regions of homogeneous high signal (*).

presented in Fig. 28-4. The depth from the skin surface of a lymphatic channel can be measured by first using the three-dimensional cursor to localize a particular lymphatic channel. An axial MPR is then created at this location, and the depth from the skin is measured. A composite postprocessed image can then be created with a full-volume MIP or volume-rendered reconstruction to provide a summary of the examination findings that show the spatial and depth information used by the surgeon for operative planning10 (Fig. 28-5). In addition to lymphatic channels, the presence, location, and size of the areas of dermal backflow are also reported9,11 (see Fig. 28-1). After the intracutaneous injection of MR contrast, contrast uptake by the veins also occurs.11 Therefore it is important to distinguish between the enhancing subdermal lymphatics and subdermal veins. Several morphologic features can be used to differentiate the two. Subdermal lymphatic channels vary in caliber, appear irregular and beaded, and are often twisted and discontinuous. In contrast, subdermal veins appear smooth with a uniform caliber and straight longitudinal course (Fig. 28-6).9,14,17 Contrast enhancement kinetics is another feature that can help to differentiate subdermal lymphatics from adjacent veins. On dynamic MRL, lymphatic enhancement generally increases over time and the enhancement progresses proximally, whereas venous enhancement peaks and then decreases over time16 (Fig. 28-7). Despite these features, it can still be difficult to determine whether a small enhancing structure is a lymphatic channel or a vein. In these cases, comparing an image location from the MRL with the same image location from the delayed MR venogram can clarify whether the structure of interest is a lymphatic channel (Fig. 28-8).

K23349_Neligan_28_Mag Reso_r4_cl_379-392.indd 386

5/28/2015 1:27:16 PM

Chapter 28  Magnetic Resonance Lymphangiography

A

387

B

C

D

FIG. 28-5  Creation of composite images for operative planning of lymphaticovenular bypass surgery. A and B, Image postprocessing algorithms are used to create full-volume MIP and volume-rendered images of the MRL of a left lower extremity in a patient with lymphedema. These measurements can then be included on the MIP and volume-rendered images to depict the location, course, and depth of a lymphatic channel on a summary image for review by the surgeon. C and D, Interactive creation of axial MPR image reconstructions from the MRL allows measurement of the depth of each lymphatic channel from the skin surface.

FIG. 28-6  The MRL appearance of enhancing lymphatic channels in a lymphedematous limb. Sagittal full-volume MIP reconstruction of the MRL of a 45-year-old woman with left leg swelling after treatment for gynecologic malignancy. On MRL, subdermal lymphatic channels appear irregular and beaded in appearance, vary in caliber, and are often twisted and discontinuous (arrows). In contrast, subdermal veins are smooth and have a uniform caliber (arrowheads).

K23349_Neligan_28_Mag Reso_r3_dc_379-392.indd 387

5/27/2015 9:33:46 AM

388

Part IV  Diagnosis of Lymphedema

FIG. 28-7  Differentiating subdermal lymphatics from veins. Coronal full-volume MIP of the MRL of a lymphedematous right arm at 5 minutes after intracutaneous contrast administration compared with 30 minutes after contrast administration showing the difference in the enhancement of the subdermal lymphatic channels and veins with time. On dynamic MRL, the beaded and irregular lymphatic enhancement (arrows) increases in intensity and progresses proximally with time, whereas the intensity of venous enhancement (arrowheads) decreases.

A 5 minutes

30 minutes

FIG. 28-8  Differentiating subdermal lymphatics from veins. Viewing the same location on the MRL and MR venogram can confirm if the structure of interest represents an enhancing lymphatic or vein. A, Coronal full-volume MIP of the MRL of a right arm shows several enhancing branching structures in the lateral forearm (arrows). Note areas of high signal (*) reflecting incomplete fat suppression. B, These enhancing structures are not present on the coronal full-volume MIP of the delayed MR venogram, indicating that they represent lymphatic channels.

B

*

*

Future Directions One of the strengths of MRL is the modality’s versatility resulting from the number of available contrast agents with different physiochemical properties and the wide array of modifiable pulse sequences that can be used to alter tissue contrast. Currently nine gadolinium-based MR contrast agents are clinically available for human use.21 Although MRL has been primarily performed with extracellular fluid agents, several of the contrast agents have specific properties that could improve MRL. Gadobutrol is an extracellular fluid agent formulated with a higher molarity (1.0 mol/L),21 which allows smaller intracutaneous injection volumes to improve patient comfort.14

K23349_Neligan_28_Mag Reso_r3_dc_379-392.indd 388

5/27/2015 9:33:46 AM

Chapter 28  Magnetic Resonance Lymphangiography

389

Based on work with different colloid-bound radiotracers used for nuclear medicine lympho­ scintigraphy,7 gadolinium chelates that associate with macroproteins22 could improve the specificity of lymphatic uptake of MR contrast. The results of a recent animal study suggested this potential benefit of protein binding for lymphatic imaging by premixing gadofosveset with human serum albumin before intradermal injection in nude mice and monkeys.23 Current clinical assessment tools used to evaluate lymphedema have significant limitations that include the lack of standardized diagnostic thresholds, high interobserver and intraobserver variation, and the inability to detect subclinical disease.24 Thus newer diagnostic techniques are needed to improve the standardization, staging, and early diagnosis of lymphedema. The ability to detect subclinical lymphedema when there is impaired lymphatic transport but no clinically evident extremity swelling would enable the early initiation of low-cost conservative treatment to maintain limb volumes and prevent disease progression.25 Because lymphatic channels in a healthy limb are typically not visualized (Fig. 28-9) on MRL, the presence of any enhanced lymphatic channels or dermal backflow indicates lymphatic dysfunction and the presence of subclinical disease.17 Quantitative information, such as lymphatic flow velocities, can be measured with dynamic contrast-enhanced MRL, potentially providing a means of noninvasively grading the degree of impaired lymphatic transport.17 Noncontrast estimates of lymphatic flow velocities are also possible,26 which is an attractive feature for surveillance programs because there is no need for intradermal contrast injections.

B

C

A

FIG. 28-9  This 74-year-old woman complained of right axillary fullness after right mastectomy. A, Axial ssFSE T2 image without fat suppression shows the fiducial marker in the right axilla placed at the site of concern indicated by the patient. B, Coronal full-volume MIP reconstruction of the T2 sequence reveals no epifascial edema in the proximal arm or axilla. The clustered foci of increased T2 signal (arrows) represent fluid within the joint spaces of the shoulder and elbow. C, Coronal full-volume MIP reconstruction of the MRL shows no enhancing lymphatic channels. The axillary fullness was the result of redundant subcutaneous soft tissue and not lymphedema. Lymphaticovenular bypass was not considered, and the patient was scheduled for cosmetic excision.

K23349_Neligan_28_Mag Reso_r3_dc_379-392.indd 389

5/27/2015 9:33:46 AM

390

Part IV  Diagnosis of Lymphedema

B

C

D

A

FIG. 28-10  This 54-year-old woman had primary lymphedema and bilateral lower extremity swelling since she was a teenager. A, Coronal full-volume MIP reconstruction of the T2 sequence of the lower legs shows severe diffuse epifascial edema symmetrically involving both lower extremities. B, A volume-rendered image of the right leg depicting the skin shows severe enlargement of the lower extremity. C, Diffuse reticular enhancement of the skin on the full-volume MIP of the MRL suggests skin thickening and fibrosis. D, Corresponding coronal MPR image of the right leg shows diffuse hypertrophy of the subcutaneous fat but no enhancing lymphatic channels. The patient was not considered a candidate for lymphaticovenular bypass.

Different MR pulse sequences can be included in a MRL examination to better characterize the subcutaneous tissues of an extremity.27 An evaluation of the soft tissue of a lymphedematous limb could differentiate early stages of lymphedema that are fluid dominant from later nonpitting solid stages, in which the increased extremity volumes are primarily from the deposition of excess adipose and fibrosclerotic tissue (Fig. 28-10). This type of disease stratification has been used to triage patients for different surgical treatment options, in which vascularized lymph node transfer and lymphaticovenular anastomosis are performed in patients with fluid-phase disease and lipectomy in patients with solid-phase chronic disease.28

Conclusion MRL is a noninvasive imaging technique that can depict individual lymphatic channels and dermal backflow in patients with lymphedema. Currently MRL is used to noninvasively diagnose and assess the severity of lymphedema, facilitate the selection of patients for microsurgery, and evaluate the response to treatment. With the increasing use and availability of microsurgical treatment for

K23349_Neligan_28_Mag Reso_r3_dc_379-392.indd 390

5/27/2015 9:33:46 AM

Chapter 28  Magnetic Resonance Lymphangiography

391

lymphedema, imaging techniques are needed to provide more accurate staging and classifying of the disease to allow earlier initiation of treatment and better individualization of treatment options. Because of the versatility of the modality, MRL has the potential to also provide functional and quantitative information about the lymphatic system, which could further improve the noninvasive assessment of lymphatic disease.

C linical P earls • Currently MRL can be used to diagnose lymphedema, plan lymphatic microsurgery, and evaluate response to treatment. • Three-dimensional image acquisition and image postprocessing techniques facilitate the interpretation of the MRL examination and allow the creation of composite images to convey spatial and depth information to the surgeon for preoperative planning. • The versatility of MR image contrast mechanisms and the clinical availability of MR contrast agents with different physiochemical properties hold the promise of further improvements in our ability to image the lymphatic system and obtain structural, functional, and quantitative lymphatic information.

R EFERENCES 1. Warren AG, Brorson H, Borud LJ, et al. Lymphedema: a comprehensive review. Ann Plast Surg 59:464472, 2007. 2. International Society of Lymphology. The diagnosis and treatment of peripheral lymphedema: 2013 Consensus Document of the International Society of Lymphology. Lymphology 46:1-11, 2013. 3. Cormier JN, Rourke L, Crosby M, et al. The surgical treatment of lymphedema: a systematic review of the contemporary literature (2004-2010). Ann Surg Oncol 19:642-651, 2012. 4. Lohrmann C, Felmerer G, Foeldi E, et al. MR lymphangiography for the assessment of the lymphatic system in patients undergoing microsurgical reconstructions of lymphatic vessels. Microvasc Res 76:4245, 2008. 5. Koshima I, Narushima M, Yamamoto Y, et al. Recent advancement on surgical treatments for lymphedema. Ann Vasc Dis 5:409-415, 2012. 6. Chang DW, Suami H, Skoracki R. A prospective analysis of 100 consecutive lymphovenous bypass cases for treatment of extremity lymphedema. Plast Reconstr Surg 132:1305-1314, 2013. 7. Szuba A, Shin WS, Strauss HW, et al. The third circulation: radionuclide lymphoscintigraphy in the evaluation of lymphedema. J Nucl Med 44:43-57, 2003. 8. Liu NF, Lu Q, Liu PA, et al. Comparison of radionuclide lymphoscintigraphy and dynamic magnetic resonance lymphangiography for investigating extremity lymphoedema. Br J Surg 97:359-365, 2010. 9. Notohamiprodjo M, Weiss M, Baumeister RG, et al. MR lymphangiography at 3.0 T: correlation with lymphoscintigraphy. Radiology 264:78-87, 2012.

K23349_Neligan_28_Mag Reso_r3_dc_379-392.indd 391

5/27/2015 9:33:46 AM

392

Part IV  Diagnosis of Lymphedema

10. Lam DL, Mitsumori LM, Neligan PC, et al. Pre-operative CT angiography and three-dimensional image post processing for deep inferior epigastric perforator flap breast reconstructive surgery. Br J Radiol 85:e1293-e1297, 2012. 11. Lohrmann C, Foeldi E, Bartholomae JP, et al. Gadoteridol for MR imaging of lymphatic vessels in lymphoedematous patients: initial experience after intracutaneous injection. Br J Radiol 80:569-573, 2007. 12. Cohan RH, Leder RA, Herzberg AJ, et al. Extravascular toxicity of two magnetic resonance contrast agents. Preliminary experience in the rat. Invest Radiol 26:224-226, 1991. 13. Ruehm SG, Schroeder T, Debatin JF. Interstitial MR lymphography with gadoterate meglumine: initial experience in humans. Radiology 220:816-821, 2001. 14. Lohrmann C, Foeldi E, Langer M. Indirect magnetic resonance lymphangiography in patients with lymphedema preliminary results in humans. Eur J Radiol 59:401-406, 2006. 15. Lohrmann C, Foeldi E, Langer M. Assessment of the lymphatic system in patients with diffuse lymphangiomatosis by magnetic resonance imaging. Eur J Radiol 80:576-581, 2011. 16. Lohrmann C, Foeldi E, Speck O, et al. High-resolution MR lymphangiography in patients with primary and secondary lymphedema. AJR Am J Roentgenol 187:556-561, 2006. 17. Liu NF, Lu Q, Jiang ZH, et al. Anatomic and functional evaluation of the lymphatics and lymph nodes in diagnosis of lymphatic circulation disorders with contrast magnetic resonance lymphangiography. J Vasc Surg 49:980-987, 2009. 18. Lu Q, Xu J, Liu N. Chronic lower extremity lymphedema: a comparative study of high-resolution interstitial MR lymphangiography and heavily T2-weighted MRI. Eur J Radiol 73:365-373, 2010. 19. Lebowitz JA, Rofsky NM, Krinsky GA, et al. Gadolinium-enhanced body MR venography with subtraction technique. AJR Am J Roentgenol 169:755-758, 1997. 20. Laurent S, Elst LV, Muller RN. Comparative study of the physicochemical properties of six clinical low molecular weight gadolinium contrast agents. Contrast Media Mol Imaging 1:128-137, 2006. 21. Mitsumori LM, Bhargava P, Essig M, et al. Magnetic resonance imaging using gadolinium-based contrast agents. Top Magn Reson Imaging 23:51-69, 2014. 22. Liu NF, Lu Q, Jiang ZH, et al. Anatomic and functional evaluation of the lymphatics and lymph nodes in diagnosis of lymphatic circulation disorders with contrast magnetic resonance lymphangiography. J Vasc Surg 49:980-987, 2009. 23. Nakajima T, Turkbey B, Sano K, et al. MR lymphangiography with intradermal gadofosveset and human serum albumin in mice and primates. J Magn Reson Imaging 40:691-697, 2014. 24. Shah C, Vicini FA. Breast cancer-related arm lymphedema: incidence rates, diagnostic techniques, optimal management and risk reduction strategies. Int J Radiat Oncol Biol Phys 81:907-914, 2011. 25. Stout Gergich NL, Pfalzer LA, McGarvey C, et al. Preoperative assessment enables the early diagnosis and successful treatment of lymphedema. Cancer 112:2809-2819, 2008. 26. Rane S, Donahue PM, Towse T, et al. Clinical feasibility of noninvasive visualization of lymphatic flow with principles of spin labeling MR imaging: implications for lymphedema assessment. Radiology 269:893-902, 2013. 27. Lohrmann C, Foeldi E, Langer M. MR imaging of the lymphatic system in patients with lipedema and lipo-lymphedema. Microvasc Res 77:335-339, 2009. 28. Granzow JW, Soderberg JM, Kaji AH, et al. An effective system of surgical treatment of lymphedema. Ann Surg Oncol 21:1189-1194, 2014.

K23349_Neligan_28_Mag Reso_r3_dc_379-392.indd 392

5/27/2015 9:33:46 AM

Part V

Current Treatment of Lymphedema

K23346_Neligan_29_Conserv Treat_r3_dc_0393-0406.indd 393

5/26/2015 9:58:45 AM

K23346_Neligan_29_Conserv Treat_r3_dc_0393-0406.indd 394

5/26/2015 9:58:45 AM

C hapter 29 Conservative Treatments for Lymphedema Neil B. Piller

K ey P oints

Con

• The treatment of lymphedema should be holistic, regardless of the area of the body affected. • The major traditional treatments improve lymphatic vessel function, soften any fibrotic tissues, reduce increased levels of connective tissues, and improve the skin as a barrier. • The mainstays of these treatments are manual lymphatic drainage (MLD) and compression bandages and garments. • Divergent opinions exist regarding the best treatment options. • An accurate assessment of the lymphedema must be made so any treatment program can be targeted and sequenced accordingly. • Many innovative treatments and management strategies have been developed that can enhance the more traditional treatment regimens.

As more is known about the lymphatic system and the effects of pathologies and surgical and radiologic disruptions on it, it is clear that various treatment and management strategies, including a multidisciplinary approach, are needed to rapidly and effectively resolve these issues and return the limb and patient to as normal as possible in a cost-effective and timely manner. The treatment of lymphedema and its comorbidities and sequelae, whatever the cause or form, has developed into a multisystem support strategy, or what was called comprehensive medical care by von Winiwarter in 1892.1 Although the names vary, depending on the group and country, lymphedema treatment is now commonly called complete decongestive physiotherapy (CDP) or complete decongestive treatment (CDT). The objectives of lymphedema treatment are holistic and include the following goals: • Improve lymphatic vessel function • Soften any fibrotic tissues • Reduce increased levels of connective tissues to normal or near-normal levels • Improve the skin as a barrier 395

K23346_Neligan_29_Conserv Treat_r3_dc_0393-0406.indd 395

5/26/2015 9:58:45 AM

396

Part V  Current Treatment of Lymphedema

Evidence for Treatment Many treatments and strategies are available for lymphedema management. For some of these treatments, there is neither little evidence of their clinical, practical biologic, or statistical effectiveness nor a clear rationale for their use, given the presenting problems. However, there is reasonable experience and an expertise base supporting most treatments. We must continue to improve our knowledge and awareness of the plethora of available treatments, and we must support attempts to gain experimental and clinical research evidence of their effectiveness so we have a better idea of what to expect in patient outcomes. The apparent lack of benefit of some of the core treatments in randomized controlled trials, metaanalysis, and systematic reviews can be daunting to a therapist or clinician. However, lymphedema is frequently a complicated pathophysiologic change to a variety of tissues and systems, and each patient is unique despite the common sign of swelling of a limb or other section of the body. The most effective way to decide on a course of treatment may be for the clinician to weigh the evidence and information and to review consensus and best practice documents to determine where the patient fits in. Different treatment options can be initiated to see which work, and targeted, sequenced treatment can proceed. In the early investigative stages of many new treatments, the clinical trials are often small, but many have a good design, objectivity, and a rigorous evaluation. Thus we can have confidence in the outcomes of such investigations. In addition to recognizing what treatments are available for a particular patient, it is important to obtain an accurate diagnosis and to know which patient to select for which treatment and the staging of these treatments. (These topics are covered in Part IV: Diagnosis of Lymphedema.) Moreover, it is very clear from a review of the common core treatments of MLD and compression that a significant proportion of poor outcomes may result from an initial poor diagnosis of the underlying problem (the swelling is not always pure lymphedema) and perhaps poor targeting and sequencing of treatment. Also, there are often different techniques and strategies to determine the presentation, status, and staging of the condition.

Traditional Conservative Treatments Manual Lymphatic Drainage The first component of CDP or CDT is a massage technique commonly called manual lymphatic drainage (MLD), of which there are many variations. Four key international variations are the Vodder technique,2 the Földi method (based on Vodder),3 the Leduc method,4 and the CasleySmith method.5 A plethora of articles have been published on the effectiveness (or lack of effectiveness) of these methods, but to date no one has compared these methods. In today’s environment the treatment that a patient with lymphedema (or at least one who is identified as being at risk for lymphedema) receives depends on the training of the therapists.

K23346_Neligan_29_Conserv Treat_r3_dc_0393-0406.indd 396

5/26/2015 9:58:45 AM

Chapter 29  Conservative Treatments for Lymphedema

397

Lasinski et al6 showed that CDT as a bundled intervention and the individual component of MLD and compression bandages is beneficial. However, a meta-analysis by Huang et al7 indicated no or little effect of MLD, throwing doubt into the minds of consumers and practitioners alike. Why this variation in outcomes and uncertainty about treatment? Most seem related to issues of methodology and rigor, but occasionally the variation results from the small size of the study groups. Although the confusion and uncertainty can be unsettling, selecting the best treatment must be dealt with, and this is where “best practice” and “templates for practice”8 and consensus documents are useful, such as that of the International Society of Lymphology,9 representing collective opinion from experience and based on individual and collective expertise.

Compression The second component of CDP is compression. The major goal of compression is to reduce venous pressure and flow and thus outflow into the interstitium, which reduces lymphatic load and encourages uploading into the lymphatics by virtue of tissue pressure variations linked to any form of muscle movement. Compression is initially delivered by bandages and then by garments in later stages (called the maintenance phase by some). Compression is usually applied with short-stretch bandages that maintain good pressure and pressure variation on movement. Equal pressure is provided at a given level of the limb by a range of padding; there is less pressure on a flat surface compared with a curved one, and stronger curves have higher pressure. A pressure gradient is established distally to proximally by careful bandaging by the therapist and at times with garment pressure monitoring. Again, benefits vary; for example, a recent trial outcome showed no benefit to below-theknee bandaging in reducing the incidence of lymphedema, whereas other studies have generally supported its benefits, with some reservations. There are summary best practice and consensus documents from the groups previously indicated.6,10-12 Furthermore, groups such as the National Lymphedema Network and the International Union of Phlebology offer specific advice on compression to help practitioners with these issues.13,14 The other way to deliver compression is through intermittent compression therapy (IPC). Although IPC has shown some benefit, its use remains controversial. Feldman et al15 stated, “In select patients, IPC use may provide an acceptable home-based treatment modality in addition to wearing compression garments.” Shao et al16 indicated that although IPC could alleviate lymphedema, there was no statistically significant difference between routine management (taken to mean best practice) of lymphedema with or without a pneumatic pump. However, within these divergent viewpoints, some trials have had good outcomes, but the results are not always consistent.

Contemporary Conservative Treatments In addition to traditional conservative treatments, many patients seek contemporary treatments, and it is important for clinicians and therapists to be aware of the range of other options for a patient with lymphedema. It is also important that the clinician and therapist are aware of any comparative benefits of these therapies. This section presents some contemporary treatments for lymphedema with which the specialist, therapist, and patient may be confronted. Some treatments are patient based with minimal or no therapist or clinical input (although this must always be en-

K23346_Neligan_29_Conserv Treat_r3_dc_0393-0406.indd 397

5/26/2015 9:58:45 AM

398

Part V  Current Treatment of Lymphedema

couraged so that there is good continuity of care), and some are administered by a lymphedema therapist or clinician. Contemporary treatments can be broadly categorized as follows: • They vibrate the tissues , encompassing a range of frequencies and amplitudes and durations. • They involve a pharmacologic agent that induces or promotes a biologic event (presented in more detail in Chapter 30). • The treatment electrically stimulates the tissues and the lymphatics. • They vary tissue pressures, including exercise (really a form of vibration). • The treatment encourages diet change and weight management. • These treatments are perhaps best described as the “placebo effect.” It should be emphasized that just because a treatment is not included here does not mean it does not work, and just because one is mentioned does not mean it is the best therapy of the group. Figures and graphs are not presented here, because these data are available in the articles and critical reviews in this chapter’s references.11 This section will describe the available treatments, the results of clinical studies, and what reasonable outcomes might be expected.

Electrostimulation Lymphatic vessels pulsate between 6 and 10 times per minute. We have shown that electrical stimulation reduces limb volume compared with the current best practice self-management group.17 Pain, heaviness, tightness, and perceived leg size (all of which are very important to the patient because they affect activities of daily living and quality of life) also improved. Other units, such as the “circulation booster,” which delivers electrical stimulation, similar in function and principle to a transcutaneous electrical nerve stimulation (TENS) unit, have also been shown anecdotally to reduce lymphedema, but trials are still in progress. Electrical stimulation of the lymphatics and surrounding skeletal muscles is likely to hold great benefit in helping lymph flow in patients with motor neuron diseases and disorders and in patients with paraplegia or hemiplegia resulting from a motor vehicle accident.

Impact of Medications on the Lymphatic System The flavonoid and benzopyrone groups are the best known group of medications that have an impact on the lymphatic system, directly or indirectly. A range of studies have been conducted on these medications, but perhaps those of Pecking et al,18 who first investigated Daflon, and Cluzan et al,19 who studied Cyclo 3 Fort, are the most representative trials. In both of these trials there were significant objective improvements in the patient groups receiving these medications. Two studies20,21 showed that another medication, Lodema, produced good outcomes in reducing patients’ lymphedema, but another study22 found that Lodema had little objective effect. Maybe this was the result of the later finding that coumarin (5,6 benzo-[alpha]-pyrone), which is the active component of Lodema, had a hepatotoxic effect in some patients. We are now aware that this serious side effect resulted from a genetic issue that affects the metabolism of coumarin. Because of our improving genetic and genomic knowledge, we can undertake genetic screening to eliminate the potential for this adverse outcome and determine who will respond well (and who may have an adverse outcome) not only to coumarin but also to the range of pharmacologic interventions

K23346_Neligan_29_Conserv Treat_r3_dc_0393-0406.indd 398

5/26/2015 9:58:45 AM

Chapter 29  Conservative Treatments for Lymphedema

399

aimed at improving lymphatic structure or function. (Further information on these and other pharmacologic interventions is detailed in Chapter 30.)

Low-Level Scanning and Handheld Laser and Light Devices Although there have been many reported case studies, the first trial with a low-level laser specifically targeting lymphedema was reported in 1993.23 According to the work of Huang et al,24 the dose per treatment area and the timing between those treatments are crucial. A double-blind, crossover, placebo-controlled trial indicated subjective and objective outcomes when low-level treatments were spread over several months.25 A continual problem in other trials and general treatment philosophies has been a lack of thought in the targeting and sequencing of treatments. I think this is one of the key reasons for the often reported poor outcomes not only for laser treatments but for treatment in general. For instance, in trials with low-level scanning and a handheld laser device, the best overall results were achieved when low-level laser therapy was used to target existing fibrotic induration of the tissues, most often associated with surgical/radiotherapeutic scarring. This therapy not only resulted in a softening of the tissues, but also a reduction in limb swelling, an improvement in in scar mobility, and generally an improvement in how the whole limb feels.26 More substantial evidence of the effects of laser therapy is still being accumulated.27 As previously mentioned, the optimal treatment time is generally short (associated with low-dose delivery), with gaps between treatments to allow the treatment effect to develop.28

Lymphatic Drainage Massage There is increasing evidence indicating the effectiveness of lymphatic drainage massage, often simple effleurage to improve lymphatic drainage when administered by appropriately trained lymph therapists.29 However, an appropriately trained patient and/or partner or caregiver can also achieve good results in between patient visits to a therapist. For this reason it is important that therapists, clinicians, and patients are aware of what they can do to improve lymphatic drainage by using either a simplified version of therapeutic massage or a range of tools that mimic the effect of massage. This can be done by changing tissue pressures and thus helping lymph move from higher pressure areas to lower ones and by recognizing that these techniques and strategies can be performed at home. Piller et al30 showed that when partners or caregivers received 10 hours of training by lymphedema therapists in the physical therapy department of a public hospital, the objective and subjective results in limb fluid reduction and perception of how the limb felt were similar to the results of professional treatment programs. One reason for this may be that the partner or caregiver knows the patient’s limb well and is acutely aware of when it is and is not responding, and with immediate feedback from the patient, the caregiver can try something different to obtain a good outcome. Of course, these programs need further research, but the positive side is that these home-based strategies not only empower the patient, but significantly reduce costs, travel time (especially for those in rural and remote areas in which therapists are often unavailable), and reestablish a “touching relationship,” which is often lacking in this older age group.

K23346_Neligan_29_Conserv Treat_r3_dc_0393-0406.indd 399

5/26/2015 9:58:45 AM

400

Part V  Current Treatment of Lymphedema

Many other home-based management strategies simply vary tissue pressures and thus help load the lymphatic system, which aids lymph flow. This category includes many massage pads, massaging beds, and handheld units, but only a few clinical trials have been done with these devices. In one of our trials we placed a midsize massage pad on patients’ lymphedematous legs. The massage pad was used in a way that was similar to how therapists work on the lymphatic drainage, which is to first clear the most proximal lymph territories, as indicated in lymphatic drainage massage programs.31 This resulted in good reductions in limb volumes, improvement in tissue softness, and patient perception of how the limb felt. It is particularly important that patients felt more in control of their condition and better able to undertake their activities of daily living. We obtained similar results with a handheld massage unit.32 Another strategy that also varied tissue pressures was passive movement of the limbs from side to side at about three times per second.33 This is really a form of vibration, and the results were similar to those of the massage pad trial in that the limb size was reduced, limbs had less fluids and were softened, and patients felt better. In these trials patients generally felt in control and were better able to undertake their activities of daily living, which is a common emerging theme linked to home-based treatment programs.

Exercise Lymphedema has many causes, and it is relatively clear that the best type of exercise for an individual depends on the severity and cause of the lymphedema and any coexisting medical conditions (for instance, arthritis, heart disease, venous disease, and diabetes). Research about the impact of exercise on breast cancer–related lymphedema is more common than for other forms of cancer, such as those occurring as sequelae of treatment of the bowel, prostate cancers, and melanomas. Exercise can be broadly grouped into the following three areas: (1) lymphedema remedial exercise, such as Tai Chi, Qigong, and other slow nonresistance programs, (2) flexibility or stretching exercises, and (3) resistance or weight-lifting exercise. The American National Lymphedema Network has published an excellent, well-balanced position paper suitable for both professionals and patients, and I recommend that the physician and patient review this article before beginning any exercise program.34 The Step-up group also has high-quality materials available.35 It is clear, however, that exercise is generally beneficial and unlikely to be injurious or to exacerbate the lymphedema. Exercise also works well with a range of other conservative treatments.36

Mild/Remedial Exercise Tai Chi and the often associated Qigong are ideal forms of exercise because patients can perform them easily and wherever and whenever needed. Tai Chi and Qigong generally vary tissue pressures and combine this with associated variations in intrathoracic and intraabdominal pressures to help facilitate the loading and flow of the lymphatic system. Patients with arm lymphedema who used a 10-minute program daily for 4 weeks achieved reductions often in the same range as more demanding treatments. Moreover, they maintained the reductions.37

K23346_Neligan_29_Conserv Treat_r3_dc_0393-0406.indd 400

5/26/2015 9:58:45 AM

Chapter 29  Conservative Treatments for Lymphedema

401

Moderate Exercise A common question from patients is, how much exercise can I do? Most studies and reviews indicate that mild exercise is good for lymphatic loading and transport because of the variation in tissue pressures that it causes. However, as the intensity of exercise increases, the surgeon must know how much of an increased lymph load a damaged lymphatic system can take. Achieving the right balance is very important, and it is essential to ensure that the patient does not have a lymphatic load that is greater than the transport capacity for a given time. The warm-down stage is the most important. Many specific exercise classes are available to patients with lymphedema of the arms and legs. Casley-Smith and Casley-Smith5 suggested that self-massage routines can be interspersed with the use of gentle movement, deep breathing, and slow rhythmic exercise of the proximal and distal muscles of the arm. Bracha and Jacob38 showed that this program reduces limb volume and improves quality of life scores. A range of water-based programs is a good way to undertake exercise with simultaneous tissue support from external pressure. An increasing number of studies provide good evidence of this.39 Water temperature is possibly an important confounder, and there remains some debate. However, Johannson and Piller40 have suggested a water temperature of 28° C (82° F). Weightlifting is a more intense exercise and encourages greater use of the limbs. One trial had patients with arm lymphedema undertake increasing levels of weightlifting while they performed a specific series of exercises to determine their limits of maximum exercise that did not worsen their lymphedema.41 Although most patients had a slight increase in limb volume immediately after exercise, the effect was short-lived as long as patients resumed their activities of daily living. This and other studies indicated that patients can undertake significant and even strenuous amounts of exercise and activity without their lymphedema worsening. However, knowledge of the limit of exercise and staying below that limit are crucial. The National Lymphedema Network has a well-researched position paper on this subject.34 The impact of exercise and significant activity on lymphedema has been reviewed, and overall there is a positive impact, varying in magnitude between studies, on limb size, range of movement, muscle strength, subjective limb symptoms, and quality of life.42 All studies advocate a cool-down period. No matter the level, exercise should be an essential part of the holistic management of a patient with lymphedema.43

Tissue Manipulation Endermologie, a technique originating in France, is used in the treatment of cellulite and similar conditions. Given the strong similarity between midstage lymphedema, cellulite, lipedema, and obesity (the adipose connection), it is also likely that Endermologie offers benefits in the treatment of lymphedema. In fact, a single-blind, randomized study of secondary arm lymphedema that compared the Endermologie technique with traditional MLD indicated similar results for MLD, although generally over a shorter time. As has been previously shown with other forms of treatment, the endermologie type of treatment showed improvements in tissue hardness and a range of subjective indicators.44 Improved outcomes were observed when the Endermologie treatment was combined with bandaging and more time was spent on clearance of the proximal lymphatic territories of the trunk and upper parts of the limb and axillary area. This is no different than what is well known and included in CDP/CLT programs.44

K23346_Neligan_29_Conserv Treat_r3_dc_0393-0406.indd 401

5/26/2015 9:58:45 AM

402

Part V  Current Treatment of Lymphedema

Kinesio/Lymph Taping Kinesio/lymph taping improves lymph drainage by lifting the skin away from the fascia that overlies the musculature, thus presumably reducing interstitial pressures in these areas and facilitating blood and lymph flow into and along these lower pressure areas. Visualizing collectors and putting tape over them is not yet being done. We can now visualize lymphatic collectors to determine lymphatic drainage pathways and the location of functional lymphatics and areas of lympathic leakage (see Chapter 27). In the future the lymphatic collectors will be visualized with indocyanine green and the tape will then be applied over them to ensure optimal lowering of pressure. Various forms of taping have been widely used in sports injuries, but recently taping has been used to treat lymphedema. Taping can be especially beneficial when garments or bandaging and other strategies are not well tolerated in hot or humid climates.45 A review of the use of the Kinesio/lymph tape for edemas indicated that the Kinesio tape was used in lymphedema management, particularly in the more challenging areas, such as the breast or chest, where bandaging and garments are difficult to use because of surface irregularities, thereby creating large, often unwanted, pressure variations.46 A trial from my clinic involving seroma after axillary clearance showed that there were significant benefits to Kinesio taping, including a reduction in the severity and duration of the seroma and the associated burning sensations, tightness, and heaviness.47 This indicates that Kinesio taping also has a role in the prevention of lymphedema by reducing the lymphatic overload and in the management of the condition.

Diet and Abdominal Issues There are many debates in the literature about diet. Generally, the human diet has gradually shifted from an antiinflammatory diet with a reasonable balance of omega-3 short-chain and omega-6 long-chain fats to a more proinflammatory diet with often up to 30 times more omega-6 than omega-3. In addition, longer chain fats must be absorbed (as chylomicrons) by way of the mesenteric lymphatics, thus placing an additional load on these lymphatics. If the latter’s structure or function is compromised in any way, such as by prior peritonitis, radiotherapy, or surgery to the area, then this absorbed load of fats may find its way into other organs or structures, particularly the groin and legs through retrograde flow.3 It is thought that a reduction of long-chain triglycerides and replacement with mid-chain and short-chain triglycerides will reduce the incidence of this retrograde flow, known as chylous reflux. Several diets based on increasing mid-chain and short-chain triglycerides have been suggested. The scientific evidence of their benefit is not strong but is improving. However, the “gray literature” has a larger amount of information. Other issues of dietary fats, pro versus antiinflammatory diets, the timing and size of meals, and the impact of inflammatory bowel disease and its effects on bloating and constipation have appeared in the gray literature and make sense from the viewpoint of minimizing or managing them. Management of constipation may reduce external pressure on the abdominal collectors as they pass to the exit point of the thoracic duct at or near the junction of the subclavian and jugular areas of the left shoulder. This pressure, which if it persists, will slow lymph flow from the legs and abdominal area and may, if prolonged and significant, result in chylous reflux into the reproductive system or legs. However, currently there is little evaluation of these effects in the scientific literature.

K23346_Neligan_29_Conserv Treat_r3_dc_0393-0406.indd 402

5/26/2015 9:58:45 AM

Chapter 29  Conservative Treatments for Lymphedema

403

Placebo In the placebo effect, a patient shows an actual or perceived improvement from a sham treatment. This effect is most likely related to the release of brain endorphins linked to the anticipation of receiving an active treatment. A systematic review has shown that placebos have a benefit in studies with continuous subjective outcome measurements, which is relevant to patients with lymphedema. Some of the trials reported in this chapter have used placebo groups. In some trials (for instance, with laser treatment),24 patients have responded to placebo treatment. This and other trials have reported an improvement in subjective symptoms.26-28 These have not always been accompanied by significant reductions in limb size or volume. An example of this is a trial investigating the effects of 5,6 benzo-[alpha]-pyrone on lymphedema after mastectomy. Here, despite an increase in physically measured arm volume in both groups, there were similar positive responses to perceived arm swelling, tightness, heaviness, and arm mobility in both the actively treated and placebo groups. As previously discussed, a handheld laser study by Carati et al,25 in which the placebo group received “sham” laser treatment for 3 weeks, followed by 1-month and 3-month follow-ups, showed that at the 3-month follow-up, the placebo group had an increase in arm volumes. However, they still reported improvement in the overall mean perceptual score and activities of daily living, much of which was statistically significant. Sometimes the placebo effect can be leveraged to the advantage of both the therapist and patient. The patient’s expectations, the therapist’s belief in the treatment being offered, and the patienttherapist relationship can contribute to the placebo effect.48,49 An awareness of these influences may help the therapist to initiate improvements in subjective symptoms, even if this is not necessarily followed by changes in other parameters such as arm volume. This relationship between the clinician and the therapy offered might be summed up by Plato, who once said, “The cure for the headache was a kind of leaf, which required to be accompanied by a charm, and if a person would repeat the charm at the same time that he used the cure, he would be made whole; but that without the charm the leaf would be of no avail.” Perhaps the telling factor is patient empowerment. Every treatment and management program must balance cost-effectiveness and benefit and whether it is in agreement with patient expectations and is also linked to any contraindications. Lymphedema requires continual diligent maintenance and effort on the part of the clinician, therapist, and patient, and any therapy that does not harm the patient and is considered beneficial is probably worth an investigation. Treatment complacency must be avoided, and perhaps changing therapy is one way to avoid this. The overarching effect of the patient’s quality of life and frame of mind may encourage the patient to undertake other treatments that will have an impact on limb size, composition, and volume. It is essential to encourage both the patient and partner/caregiver to play a role, but we should always be realistic regarding our expectations and compliance requests.50,51 Otherwise, the very person we want to help may or may not be able to comply with our instructions.

K23346_Neligan_29_Conserv Treat_r3_dc_0393-0406.indd 403

5/26/2015 9:58:45 AM

404

Part V  Current Treatment of Lymphedema

Conclusion There are many different conservative treatments, although unfortunately not all are supported by large-scale, blind, crossover trials. Quite possibly this will never occur because of the lack of financial support from manufacturing or pharmaceutical firms. However, there are some small trials of reasonable scientific merit, and an increasing body of evidence indicates that many of these conservative treatments help patients practically, even if the outcomes do not appear statistically significant. The use of some of these conservative treatments helps empower patients and allows them to see that they and their partner or caregiver can often make a significant difference in their quality of life. Most conservative treatments can be done at home, which is less expensive and less time consuming than an office visit with a therapist. Certainly any conservative treatment that is done at home should be performed with the knowledge of a trained health professional, who may also provide some form of MLD, skin care, and other recommendations. The physician should also try one treatment at a time and give that treatment sufficient time to determine whether it is effective. In this way it is similar to a randomized controlled trial of one. Only then will the physician and patient know what treatment is effective. Very few conservative treatments do harm; additional treatment or stronger treatment is not necessarily better. Slow and gentle treatment done one step at a time is preferable for patients with lymphedema.

C linical P earls • An initial holistic assessment of the patient is critical to formulate the best recommendation for treatment or management of the lymphedema. • Good treatment is targeted and sequenced according to this thorough assessment. • It is a good idea to ask patients what they want as an outcome of the intervention. Sometimes limb size is not the problem they focus on; it is how the limb feels, issues of mobility, or the ability to perform activities of daily living. • A plethora of treatments exist, and patients may bring new ones to your attention. The surgeon should know whether there is any possibility of harm, and if not, allow the patient to try the treatment and see if it is effective. • The physician should encourage patients with lymphedema to give any treatment a reasonable time to act; after all, in many cases there has been swelling for years. Patients should also be encouraged to only try or do one thing different at a time. In this way both the physician and patient can determine whether the treatment is effective. • The physician will encounter patients who already have lymphedema and patients who are at risk of lymphedema or have lymphedema in its very early stages. • The physician should carefully consider how to be proactive with new patients. The recommended treatment may make a great difference to the development of an individual’s lymphedema.

K23346_Neligan_29_Conserv Treat_r3_dc_0393-0406.indd 404

5/26/2015 9:58:45 AM

Chapter 29  Conservative Treatments for Lymphedema

405

R EFERENCES 1. von Winiwarter A. Die Elephantiasis. In Deutsche Chirurgie. Stuttgart: Enke, 1892. 2. Vodder E. Manuelle Lymphdrainage. Deutsche Badebetriebe 56:386-388, 1965. 3. Stroessenreuther RHK, Deuss J. Manual lymphatic drainage (MLD) according to Dr E Vodder. In Földi M, Földi E, Kubik S, eds. Textbook of Lymphology, ed 2. Munich: Urban & Fischer, 2012. 4. Leduc O, Leduc A. Manual lymph drainage (Leduc method). In Lee BB, Bergan J, Rockson SG, eds. Lymphedema: A Concise Compendium of Theory and Practice. London: Springer-Verlag, 2011. 5. Casley-Smith JR, Casley-Smith JR. Modern Treatment for Lymphoedema, ed 5. Adelaide, Australia: The Lymphoedema Association of Australia, 1997. 6. Lasinski BB, McKillip Thrift K, Squire D, et al. A systematic review of the evidence for complete decongestive therapy in the treatment of lymphedema from 2004 to 2011. PMR 4:580-601, 2012. 7. Huang TW, Tseng SH, Lin CC, et al. Effects of manual lymphatic drainage on breast cancer–related lymphedema: a systematic review and meta-analysis of randomized controlled trials. World J Surg Oncol 11:15, 2013. 8. International Lymphoedema Framework documents. Available at www.lympho.org/resources.php. 9. International Society of Lymphology. The diagnosis and treatment of peripheral lymphedema: 2013 consensus document of the International Society of Lymphology. Lymphology 46:1-11, 2013. 10. Stuiver M, de Rooij JD, Lucas C, et al. No evidence of benefit from class II compression stockings in the prevention of lower-limb lymphedema after inguinal lymph node dissection: results of a randomized controlled trial. Lymphology 46:120-131, 2013. 11. McNeely ML, Peddle CJ, Yurick JL, et al. Conservative and dietary interventions for cancer-related lymphoedema: a systematic review and meta-analysis. Cancer 117:1136-1148, 2011. 12. Lee BB, Bergan J, Rockson SG, eds. Lymphedema: A Concise Compendium of Theory and Practice. London: Springer-Verlag, 2011. 13. Lee BB, Andrade M, Antignani PL, et al. Diagnosis and treatment of primary lymphedema. Consensus document of the International Union of Phlebology (IUP)-2013. Int Angiol 32:541-574, 2013. 14. Stout N, Partsch H, Szolnoky G, et al. Chronic edema of the lower extremities: international consensus recommendations for compression therapy clinical research trials. Int Angiol 31:316-329, 2012. 15. Feldman JL, Stout NL, Wanchai A, et al. Intermittent pneumatic compression therapy: a systematic review. Lymphology 45:13-25, 2012. 16. Shao Y, Qi K, Zhou QH, et al. Intermittent pneumatic compression pump for breast cancer-related lymphedema: a systematic review and meta-analysis of randomized controlled trials. Oncol Res Treat 37:170-174, 2014. 17. Piller NB, Douglas J, Heidenreich B, et al. Placebo controlled trial of mild electrical stimulation. J Lymphoedema 5:15-25, 2010. 18. Pecking AP, Février B, Wargon C, et al. Efficacy of Daflon 500 mg in the treatment of lymphedema (secondary to conventional therapy of breast cancer). Angiology 48:93-98, 1997. 19. Cluzan RV, Alliot F, Ghabboun S, et al. Treatment of secondary lymphedema of the upper limb with CYCLO 3 FORT. Lymphology 29:29-35, 1996. 20. Casley-Smith JR, Morgan RG, Piller NB. Treatment of lymphoedema of the arms and legs with 5-6-benzo-[alpha]-pyrone. N Engl J Med 329:1158-1163, 1993. 21. Casley-Smith JR. Benzopyrones in the treatment of lymphoedema. Int Angiol 18:31-41, 1995. 22 Loprinzi CL, Kugler JW, Sloan JA, et al. Lack of effect of coumarin in woman with lymphoedema after treatment for breast cancer. N Engl J Med 340:346-350, 1990. 23. Piller N, Thelander A. Treatment of chronic postmastectomy lymphedema with low level laser therapy: a 2.5 year follow-up. Lymphology 31:74-86, 1998. 24. Huang YY, Chen AC, Carroll JD, et al. Biphasic dose response in low level light therapy. Dose Response 7:358-383, 2009. 25. Carati CJ, Anderson SN, Gannon BJ, et al. Treatment of postmastectomy lymphedema with low-level laser therapy: a double blind, placebo-controlled trial. Cancer 98:1114-1122, 2003.

K23346_Neligan_29_Conserv Treat_r3_dc_0393-0406.indd 405

5/26/2015 9:58:45 AM

406

Part V  Current Treatment of Lymphedema

2 6. Wigg J. Use and response to treatment using low level laser therapy. J Lymphoedema 4:73-76, 2009. 27. Lee N, Wigg J, Carroll J. The use of low level light therapy in the treatment of head and neck oedema. J Lymphoedema 8:35-42, 2013. 28. Tilley S. Use of laser therapy in the management of lymphedema. J Lymphoedema 4:39-72, 2009. 29. Williams A. Manual lymphatic drainage: exploring the history and the evidence base. Br J Community Nurs 15:S18-S24, 2010. 30. Piller NB, Packer R, Coffee J, et al. Accepting responsibility for health management. Partner training as an effective means of managing chronic lymphedema. Progress in Lymphology XC. Lymphology 29(Suppl):266-269, 1996. 31. Moseley AL, Piller NB, Carati C. A systematic review of common conservative therapies for secondary limb lymphedema: which is best and when should they be used? Lymphology 41(Suppl):531-534, 2008. 32. Piller NB, Moseley A, Heidenreich B, et al. Giving control over lymphoedema back to patients: results of a clinical trial of a cycloid vibratory hand held massage (Lymphease ) unit for chronic arm lymphedema. Lymphology 41(Suppl):519-522, 2008. 33. Piller NB, Moseley A, Esterman A, et al. The Sun Ancon Chi Machine Aerobic Exerciser: a new patient focused home based therapy for people with chronic secondary lymphedema. Lymphology 37:53-61, 2004. 34. National Lymphedema Network. Exercise: Position Statement of the National Lymphedema Network. NLN Medical Advisory Committee, updated May 2011. Available at www.lymphnet.org. 35. Step Up, Speak Out. Breast Cancer-Related Lymphedema: Essential Knowledge for Personal Trainers, Fitness Instructors, Yoga Teachers and Pilates Teachers, 2013. Available at www.stepup-speakout.org. 36. Chang CJ, Cormier JN. Lymphedema interventions: exercise, surgery, and compression devices. Semin Oncol Nurs 29:28-40, 2013. 37. Moseley A, Piller NB, Carati C. The effect of gentle arm exercise and deep breathing on secondary arm lymphedema. Lymphology 38:136-145, 2005. 38. Bracha J, Jacob T. Using exercise classes to reduce lymphedema. J Lymphedema 5:46-55, 2009. 39. Tidhar D, Katz-Leurer M. Aqua lymphatic therapy in women who suffer from breast cancer treatmentrelated lymphedema: a randomized controlled study. Support Care Cancer 18:383-392, 2010. 40. Johansson K, Piller NB. Exercises with heavy weights for patients with breast cancer related lymphoedema 2005. Proceedings of the Twentieth International Congress of Lymphology, Salvador, Brazil, Sept 2005. 41. Johansson K. Weight-bearing exercise and its impact on arm lymphoedema. J Lymphoedema 2:115122, 2007. 42. Moseley A, Piller NB. Exercise for limb lymphoedema: evidence that it is beneficial. J Lymphoedema 3:51-56, 2008. 43. Todd M. Self-management in chronic oedema. Br J Nurs 22:701-702, 2013. 44. Moseley A, Piller NB, Douglass J, et al. Comparison of the effectiveness of MLD and LPG. J Lymphoedema 2:30-36, 2007. 45. Rock-Stockheimer K. Kinesio Taping for Lymphedema and Chronic Swelling. Albuquerque, NM: Kinesio USA, 2006. 46. Finnerty S, Thomason S, Woods M. Audit of the use of kinesiology tape for breast oedema. J Lymphoedema 5:38-44, 2010. 47. Bosman J, Piller NB. A randomized clinical trial of lymph taping in seroma formation after breast cancer surgery. J Lymphoedema 5:12-23, 2010. 48. Benson H, Friedman R. Harnessing the power of the placebo effect and renaming it “remembered wellness.” Ann Rev Med 47:193-199, 1996. 49. Finniss DG, Kaptchuk TJ, Miller F, et al. Biological, clinical, and ethical advances of placebo effects. Lancet 375(9715):686-695, 2010. 50. Piller NB. We need to help consumers to help themselves. J Lymphoedema 7:6, 2012. 51. Dart L. Non-compliance? It’s easy for you to say . . . ! J Lymphoedema 7:8, 2012.

®

K23346_Neligan_29_Conserv Treat_r3_dc_0393-0406.indd 406

5/26/2015 9:58:46 AM

C hapter 30 Pharmacologic Treatment of Lymphedema Kathleen Wang

Pha

K ey P oints • When clinicians identify a patient’s nonadherence to long-term conservative therapies, such as the wearing of compression garments and manual lymphatic drainage, pharmacotherapy is an option that is often overlooked. • Although benzopyrones are not routinely used as treatment for lymphedema, many clinical studies have found that they offer therapeutic benefits. • There is a growing trend toward the use of complementary and alternative medicine as a supplement to allopathic treatments for lymphedema. This includes the use of herbal therapies as well as vitamins, with variable efficacy in symptom reduction. • Filariasis is the most common cause of secondary lymphedema; it affects 120 million people worldwide, and if left untreated, will develop into disfiguring elephantiasis. Treatment involves elimination of the infective organisms and should be initiated promptly. • Repeated bouts of lymphedema-related cellulitis may require long-term prophylactic antibiotic therapy. • In patients whose impaired lymphatic system is overwhelmed by the task of absorbing the body’s basal interstitial fluid load, the administration of certain drugs that further increase capillary filtration will worsen fluid pooling. Resultant edema can cause further damage to lymphatic structures through the dilation of these vessels. Where possible, individuals should avoid these medications. • More methodologically robust clinical trials are necessary to verify the efficacy of the various pharmacotherapies for chronic lymphedema.

The lymphatic system is a network of lymph nodes and vessels that operate in parallel with the vascular system to provide immune and transport functions; lymphedema is a chronic condition caused by an acquired or congenital defect of the lymphatic system. Normal and healthy lymphatic vessels are responsible for absorbing and then transporting excess interstitial fluid to the blood. Compromised lymphatic integrity diminishes this transport capacity, and high-protein fluids remain trapped in the interstitial spaces (principally the subcutaneous fatty tissues), which results in edema. Fluid that pools in the lymphatic vessels also causes their dilation, thereby producing incompetent lymphatic valves in the collecting ducts as well as compounded fluid sta407

K23346_Neligan_30_Pharma Treat_r3_0407-0422.indd 407

5/26/2015 10:00:14 AM

408

Part V  Current Treatment of Lymphedema

sis. This promotes a cascade of cellular events that favor chronic inflammation and pain. In addition, ongoing edema compresses the surrounding healthy lymphatic vessels, which impedes fluid flow and exacerbates regional swelling. If there are sufficient lymphatic structures to compensate for local defects, the problem remains subclinical. More commonly, however, a clinical picture of lymphedema is observed. Patients present with severely edematous limbs, poor wound healing, and acute episodes of cellulitis.1 Current best practices recommend conservative combination treatment for lymphedema,2 which is generally biphasic.3 The first “intensive” phase involves the timely provision of manual lymphatic drainage (MLD) massage by a trained service provider, instructions for therapeutic exercises, and medical-grade short-stretch compression bandages to be worn daily. This phase may last a few months, depending on the frequency of treatment sessions; its aim is to stabilize the lymphedema by preventing further damage to the lymphatic system from regional fluid overload. The subsequent “maintenance” phase lasts indefinitely. Lymphedema is a chronic and progressive condition, but it is subject to an individual’s willingness to continue with the tasks of maintenance. These tasks include wearing a well-fitted gradient pressure garment during the day and compression bandages at night, performing appropriate skin care, continuing therapeutic exercises, and selfadministering MLD.4 For some patients, these tasks represent an overwhelming ongoing burden, and they discontinue their daily efforts.5 This is understood as nonadherence. Nonadherence is a significant challenge to the management of lymphedema for both patients and clinicians. This is especially true when it comes to patients wearing compression garments (which is pivotal for controlling lymphedema progression6) and continuing with MLD treatment. There are often understandable reasons for nonadherence. Many patients with lymphedema report trouble with accessing correctly fitted garments in a timely manner. For elderly individuals and those with mobility impairment, donning such garments is a daily struggle. Individuals who live in hot, humid climates find it difficult to wear compression garments, because these will be uncomfortable in this weather, even when correctly fitted. The cost of purchasing compression garments is another barrier to continued use, especially because these garments need to be replaced frequently to remain therapeutic. Many patients also struggle to access and afford MLD, which is an ongoing cost that can make long-term treatment prohibitive.7 For cases in which there are legitimate reasons for nonadherence to conservative therapies, alternatives must be considered. Surgery is one such option, particularly if conservative treatments are adhered to but are still ineffective. Pharmacotherapy is another option, but it remains fairly overlooked in clinical practice. This chapter will provide an overview of the drugs that have been trialed for the treatment of different types of lymphedema and for the complications associated with lymphedema.

Benzopyrones Benzopyrones are considered to have antiedematous properties and have been used successfully for the treatment of certain venous diseases. There are two major groups of benzopyrones: coumarins and flavonoids.8

K23346_Neligan_30_Pharma Treat_r3_0407-0422.indd 408

5/26/2015 10:00:15 AM

Chapter 30  Pharmacologic Treatment of Lymphedema

409

Coumarins, which are alpha-benzopyrones, belong to the coumarol family; they act on vascular permeability, macrophages, and inflammatory mediators.9 Many authors advocate their use for the treatment of lymphedema because of their potential to reduce high-protein edema through multimodal mechanisms. Coumarin and its metabolites adhere to the endothelium of small blood vessels, reducing vascular permeability and decreasing protein leakage into the interstitial tissues.8 Coumarins also promote the influx of macrophages into edematous tissues, which stimulates proteolysis to reduce the accumulation of the proteins that favor the development of edema. This in turn reduces the interstitial oncotic pressure and helps with the vascular uptake of fluid to ease the lymphatic fluid overload. In patients with chronic lymphedema, macrophages can become structurally altered (they have fewer pseudopodia and are more lipid rich) and less functionally active.10 There is research to suggest that this inactivated state can be reversed by benzopyrones and that it is accompanied by a correlated elevation in proteases (typically secreted by activated macrophages).11 Coumarins also inhibit the synthesis of inflammatory mediators such as prostaglandins and leukotrienes to lessen the chronic inflammatory processes that cause irreversible tissue fibrosis.8 Finally, animal models have shown that coumarins increase lymph flow.10 This is presumably facilitated by increased lymphatic contractility and pumping, which improve lymphatic clearance through healthy neighboring lymphatic structures to reduce edema. Flavonoids are gamma-benzopyrones that include flavones; diosmin; flavan and its derivative hesperidin; quercetin and its derivatives rutin, oxerutin, and the rutosides; and proanthocyanidins. Like the coumarins, flavonoids are thought to reduce the symptomatic swelling seen in patients with lymphedema, because they reduce vascular leakage and the capillary filtration rate. Flavonoids are also antioxidant compounds with inhibitive actions that affect the enzymatic promoters of inflammation.8 However, it has been observed that larger quantities of flavonoids (compared with coumarins) need to be consumed to produce similar effects.10 Benzopyrones are not routinely used as treatment for lymphedema, but many clinical studies have found that they offer therapeutic benefits. A meta-analysis of 27 clinical trials (of varying quality) that studied the effects of oral benzopyrones on peripheral lymphedema was carried out by prominent lymphedema researchers.10 The lymphedemas studied included congenital (primary), iatrogenic (after surgery and radiotherapy), and filarial. Most trials spanned 6 months, but some lasted 1 year or more. The benzopyrones investigated included the coumarins, the oxerutins/rutosides, the troxerutins, diosmin, and hesperidin. Coumarin dosages were typically 400 mg/day (although some were as low as 100 mg/day) and produced significant improvements in edema, limb circumference, skin softness and thickness, pain, limb heaviness and mobility, frequency of acute inflammatory episodes and ulcers, and general well-being. Oxerutin trials involved the administration of 3000 mg/day and had variable results. Some studies found no significant symptomatic changes, whereas others found improvements in skin temperature, edema, limb circumference, limb mobility, pain, heaviness, and well-being. Troxerutin was only studied in combination with coumarin, and dosages varied from 60 to 270 mg/day. Several studies reported reduced limb circumference and secondary inflammation (cellulitis, lymphagitis, and lymphadenitis) as well as improved lymph flow and symptoms. A few studies observed improved lymph flow but no changes in symptoms. Finally, one study found reductions in edema and other symptoms in addition to

K23346_Neligan_30_Pharma Treat_r3_0407-0422.indd 409

5/26/2015 10:00:15 AM

410

Part V  Current Treatment of Lymphedema

improved lymph flow when patients were given a mixture of micronized diosmin (90%) and hesperidin (10%) at 1000 mg/day. The meta-analysis of the combined results of these clinical studies demonstrated that benzopyrone is effective for the reduction of limb edema and symptoms, but it also found that these effects can take at least 6 months to become clinically evident. It was also noted that higher doses were correlated with higher reductions and that the coumarins and oxerutins were superior to other benzopyrones. The results from 13 of the 16 double-blind trials showed significant symptomatic improvement, and a reduced rate of secondary inflammation was found in all 16 trials. In light of these promising results, the authors of the meta-analysis recommended the use of benzopyrones for the treatment of lymphedema. By contrast, a more recent Cochrane review found insufficient evidence to support this treatment recommendation.12 The authors believed that there were too many methodologic problems in the 15 trials that were identified to conduct a rigorous meta-analysis. Thus they were unable to conclusively comment on the efficacy of benzopyrones or to recommend their use. For clinicians who are interested in using benzopyrones to treat lymphedema, it should be noted that oral coumarins are associated with potentially fatal hepatotoxicity in up to 6% of patients13 and that they have been withdrawn from the market in both the United Kingdom and Australia.8 Topical coumarin is an alternative to the oral version, and it was demonstrated in four trials to improve swelling when used in combination with compression garments.14 It can sting if it is applied directly to the mucous membranes, but very few application-site rashes have been reported.10

Herbal Pharmacotherapies Around the world, there is a growing trend toward the use of complementary and alternative medicine (CAM) as a supplement to allopathic therapy. This includes the use of herbal therapies as well as vitamins. In Australia, nearly half of the population use CAM and annually spend approximately $2.3 billion on these products.15 MacLennan et al16 found that many CAM consumers are self-prescribers; this is particularly alarming for herbal medicines, which can have as many toxic side effects as conventional synthetic drugs. Many herbal therapies are misrepresented as safe by virtue of their being “natural,” and considerable ambiguity exists in the scientific and gray literature with regard to their efficacy. The herbs discussed in the following sections have been clinically studied. Each description details the herb’s active ingredients, its posited mechanism of action, the evidence of its efficacy (or lack thereof), and its documented contraindications and possible adverse reactions.

Horse Chestnut Seed (Aesculus hippocastanum ) The active component of horse chestnut seed extract (HCSE) is escin, a triterpenic saponin that can inhibit the activity of elastase and hyaluronidase.17 These enzymes normally degrade proteoglycan in capillary endothelium and promote vascular leakage, which can overload the lymphatic system. HCSE also increases the acid protease activity levels associated with an increased rate of edema resolution and reduced swelling. Therefore, it has potential for the treatment of lymphedema. Hutson et al18 clinically tested HCSE use by 25 breast cancer survivors with stable arm

K23346_Neligan_30_Pharma Treat_r3_0407-0422.indd 410

5/26/2015 10:00:15 AM

Chapter 30  Pharmacologic Treatment of Lymphedema

411

lymphedema. For 3 months the participants were given either the HSCE equivalent of 50 mg of escin twice daily, or placebo. Unfortunately, no significant changes in lymphedema were observed, either in terms of objective arm fluid and size measurements or the participants’ subjective ratings of improvement. Despite the lack of evidence supporting HCSE’s therapeutic benefit, some patients may still choose to try it. These patients must be warned about its side effects. Although no teratogenicity, embryo toxicity, or chronic toxic effects have been reported with HCSE use, it can cause a number of other side effects, including muscle twitching, weakness, impaired coordination, dilated pupils, dizziness, headaches, pruritus, vomiting, and diarrhea. Dosages should not exceed 600 mg/day because of the risk of nephrotoxicity. Furthermore, HCSE should not be ingested by pregnant or breast-feeding women, people with bleeding disorders or with renal or hepatic impairments, or people who are taking anticoagulants.

Butcher’s Broom (Ruscus aculeatus) The active components of butcher’s broom include the saponin glycosides of ruscogenin and neoruscogenin, which have antiinflammatory,19 antiexudative, and edema-reducing properties. To date, no trial has studied butcher’s broom extract without other additives. The most widely available form is Cyclo 3 Fort (C3F), which is a combination of butcher’s broom extract (150 mg), hesperidin methyl chalcone (150 mg), and ascorbic acid (100 mg). Cluzan et al20 administered three C3F tablets daily to 27 patients with lymphedema and found that those taking the C3F had a significantly greater total decrease in whole arm volume compared with the placebo group. This observation was true on days 30, 60, and 90 of the clinical trial. In addition, 75% of the C3F group also reported improvements in arm quality, softness, and mobility, whereas only 32% of the placebo group reported these effects; this was a statistically significant difference. However, there were problems with the study’s methodology, including reporting, attrition, and selection biases. Therefore, despite the encouraging results, the study’s validity is questionable. The researchers also noted nausea and abdominal pain in two participants, which suggests that not all individuals are able to tolerate C3F.

Ginkgo (Ginkgo biloba) The main active ingredients in ginkgo are ginkgolides and flavonoids, which are thought to protect cellular lysosomes from free radicals and to improve the integrity of collagen fibers. Cluzan et al21 studied the effect of Ginkor Fort, a ginkgo extract, on breast-cancer–related arm lymphedema. Forty-eight patients were studied, and a statistically significant improvement in limb heaviness (based on self-reported measures) was observed. However, no definitive conclusions could be drawn with regard to this herb’s effect on lymphatic migration speed, and the researchers recommended further investigation to establish efficacy.

Sweet Clover (Melilotus officinalis) and Tonka Beans (Dipteryx odorata) The primary active ingredient in both sweet clover and tonka beans is coumarin. For 6 months Pastura et al22 administered 400 mg/day of sweet clover, which contained 8 mg of coumarin, to 21 survivors of breast cancer with secondary lymphedema. The researchers concluded that sweet clover effectively reduced lymphedema in 79% of the participants treated. Unfortunately, these

K23346_Neligan_30_Pharma Treat_r3_0407-0422.indd 411

5/26/2015 10:00:15 AM

412

Part V  Current Treatment of Lymphedema

results need to be viewed with caution, because only 14 participants were treated, according to the study protocol. Although the reduction was statistically significant, it was small (about 5%). Three participants also experienced transitory gastrointestinal side effects. In another study, Vettorello et al23 studied the effect of sweet clover, tonka beans, and ginkgo combined. Seventy-six patients with lower limb lymphedema were treated in an open-label study for 6 to 8 months, and significant improvements in limb circumference, edema, episodes of infection, and limb pain and heaviness were demonstrated. However, there were methodologic problems with this study. Health care providers should be aware that coumarin can cause hepatotoxicity in people with inactive or nonfunctional cytochrome P450 2A6.24 Patients who insist on trying plant-based coumarin should undergo an assay of their cytochrome P450 2A6 function before they begin taking the drug.

Vitamins Several online lymphedema support groups advocate for the use of vitamins and minerals in managing lymphedema. The vitamins include A, B complex, C, and E and are thought to be therapeutic because of their antioxidant properties and their role in improved wound healing and immunity. The minerals include zinc, selenium, and copper. Although animal studies have shown that vitamin B complex and vitamin P (a flavonoid) are effective in reducing lymphedema,25 only selenium has undergone clinical trials for its efficacy in reducing inflammation and edema in patients with lymphedema.

Selenium Selenium is an essential trace element that is nutritionally necessary for thyroid hormone metabolism, DNA synthesis, and protection against oxidative damage and infection. With respect to lymphedema, it was hypothesized that selenium would have therapeutic benefit because of its antioxidant and free-radical scavenging properties. Selenium activates glutathione peroxidase, which is responsible for reducing free radicals and which may also have immunostimulant effects.26 Two clinical trials investigated selenium as a pharmacotherapeutic treatment for limb lymphedema. In both trials, cancer survivors with lymphedema were given 3 weeks of intensive physical therapy with either oral selenium (sodium selenite) or placebo. This was followed by a 3-month period, during which a smaller dose of selenium (or placebo) was continued. Measures of inflammation and edema were taken at 3 weeks and 3 months. In the first trial, no inflammatory episodes were reported for the selenium group at the third week and third month (n 5 19), whereas participants in the placebo group (n 5 15) had one episode of inflammation at week 3 and seven episodes by the third month.27 Similarly, in the second trial, the selenium group (n 5 32) suffered no inflammatory episodes; the placebo group (n 5 28) reported 1 and 14 episodes at the two measuring points, respectively.28 Neither trial reported any improvement in limb swelling.

K23346_Neligan_30_Pharma Treat_r3_0407-0422.indd 412

5/26/2015 10:00:15 AM

Chapter 30  Pharmacologic Treatment of Lymphedema

413

Unfortunately, there were serious methodologic weaknesses in these trials, including inadequate reporting of dosage details, the methods of randomization and blinding, and details of outcome assessments, such as the degree and duration of edema. The authors of two Cochrane reviews that addressed selenium use with lymphedema thus concluded that there is insufficient evidence to support the use of selenium supplementation as a treatment for lymphedema or its inflammatory complications.29 Bruns et al30 studied the efficacy of selenium for the treatment of head and neck lymphedema caused by radiotherapy alone or in combination with surgery. Thirty-six patients with cancer were treated with oral sodium selenite (50 mg per day) for 4 to 6 weeks after radiotherapy. Measures of edema and quality of life were taken before and after treatment, and the researchers found that selenium supplementation reduced edema and significantly improved quality of life. However, given the small sample size, further research is needed to validate these promising results before a definitive recommendation can be given. For clinicians who are interested in prescribing selenium, note that daily intake should not exceed 400 mg for adults when selenium is to be taken on a long-term basis. This is because selenosis (chronically high selenium intake) is associated with lesions of the skin and the nervous system, nausea, diarrhea, dental enamel abnormalities, brittle nails, hair loss, fatigue, irritability, and neurologic symptoms.31

Drugs for Lymphatic Filariasis Worldwide, filariasis is the most common cause of secondary lymphedema, and it affects 120 million people.32 This infection is caused by parasitic nematodes that are transmitted by female mosquitoes as larvae. Once they are inside the human host, these larvae inhabit the lymphatics and the subcutaneous tissues; this colonization is called microfilariae infection. The larvae mature in the lymphatics and become adult worms within 12 to 15 days (macrofilariae phase). They begin to reproduce 12 months after maturation, and the next-generation larvae are disseminated through the circulatory system. Most individuals are infected as children and remain asymptomatic, although they demonstrate lymphatic changes such as lymphangiectasis (dilation of the lymphatic vessels) as a result of the resident macrofilariae. For less fortunate individuals, overt lymphedema manifests with severe limb, breast, and scrotal swelling that will develop into permanently disfiguring elephantiasis if untreated. Other symptoms include fever, lymphangitis (inflammation of the lymph vessels), lymphadenopathy (enlargement of the lymph nodes), and hydroceles. Opportunistic bacterial infections can also cause cellulitis, which further damages the lymphatic system and exacerbates swelling and regional pain.33 Pharmacotherapy for lymphatic filariasis typically involves the elimination of the infective agent; this can prevent the development of chronic lymphedema if treatment is initiated promptly. Unfortunately, the drugs do not reverse the damage to the lymphatic structures. When deciding which drugs to administer, it is important to recognize the presence of concomitant infections such as onchocerciasis (also known as river blindness and Robles’ disease). This will determine which drugs to avoid to protect patients from severe adverse reactions, including blindness.

K23346_Neligan_30_Pharma Treat_r3_0407-0422.indd 413

5/26/2015 10:00:15 AM

414

Part V  Current Treatment of Lymphedema

Diethylcarbamazine (DEC) is the treatment of choice for patients with monoinfection. DEC, which is a piperazine derivate, has potent microfilaricidal and macrofilaricidal actions against Wuchereria bancrofti, Brugia malayi, and Brugia timori, possibly through the disruption of microfilarial surface membranes and enhanced host immunity. Studies estimate that DEC can kill up to 50% of adult worms, which consequently decreases the microfilarial load. DEC should be administered for 12 days (6 mg/kg/day), even when clinical symptoms are absent. This is the recommended treatment protocol, because it confers protection from—and in some instances, results in the reversal of—early pathologic lymphatic changes that can occur before symptom onset. However, it is strongly contraindicated for patients with onchocerciasis as well as those who are pregnant. Some patients experience adverse effects such as fever, headache, anorexia, nausea, and arthralgias, which can be managed with antipyretics and antiinflammatory medications.34 The addition of doxycycline (200 mg/day) for up to 6 weeks has also been recommended, because studies have demonstrated its macrofilaricidal activity. Although it provides limited clinical improvement for patients with symptoms of advanced lymphatic filariasis, doxycycline is still a recommended course of treatment. It kills remaining macrofilariae through the elimination of their bacterial symbiont, Wolbachia. A few studies have found that doxycycline, either alone or in combination with DEC, reduces lymphangiectasis and hydroceles in affected individuals. Unfortunately, doxycycline affects developing cartilage and bone, so it is contraindicated for children who are less than 8 years old as well as during pregnancy.34 Ivermectin can also be used to treat lymphatic filariasis as a result of its microfilaricidal activity. However, it does not successfully eliminate microfilariae, and it must be administered in repeated doses to kill each new generation of microfilariae. Despite this, it is the treatment of choice for patients with concomitant onchocerciasis. A single dose of ivermectin (150 mg/kg) helps to clear the Onchocerca volvulus microfilariae so that DEC can be safely given 1 month later. Note that ivermectin cannot be given to patients with loiasis (also known as loa loa filariasis or eye worm) because of a risk of posttreatment encephalopathy. For these individuals, albendazole is a viable alternative, because it targets microfilariae; this will gradually reduce the microfilariae load.34 With regard to interrupting the transmission of lymphatic filariasis, a Cochrane review concluded that there was insufficient evidence to support the efficacy of albendazole.35

Antibiotics for Lymphedema-Related Cellulitis Approximately 45% of patients with non-cancer-related lymphedema experience an acute inflammatory episode, and some struggle with recurrences every few weeks.29 Cellulitis is a major complication of lymphedema; it is clinically characterized by fever, regional pain, and skin rubor with poorly defined margins.36 Its pathogenesis is related to the anatomic changes that accompany lymphedema. Individuals with chronic lymphedema demonstrate epidermal thickening and hyperkeratosis. The sebaceous glands may also reduce their secretions, and this results in dry skin that can crack and lose its function as a first-line immune barrier.37 The skin becomes vulnerable to infectious pathogens such as streptococci and staphylococci. Specifically, group A, B, and G hemolytic streptococci organisms are most commonly associated with lymphedema cellulitis. Cases of staphylococcal and methicillin-resistant Staphylococcus aureus–related (MRSA) cellulitis have also been documented.38

K23346_Neligan_30_Pharma Treat_r3_0407-0422.indd 414

5/26/2015 10:00:15 AM

Chapter 30  Pharmacologic Treatment of Lymphedema

415

Human skin relies heavily on the lymphatic system for immune defense. Lymphatic vessels form a passage for macrophages and Langerhans cells, which gather antigens from the skin surface. When there is healthy lymphatic architecture and function, these cells are transported to the lymph nodes, where they initiate an immunologic response.39 Similarly, the clearance of bacteria from the interstitium requires healthy lymphatic vessels that pump fluid and debris. In the absence of these functional lymphatic vessels, there is inadequate immune protection against infection.40 Cellulitis requires immediate treatment; it is an aggressive inflammation that can cause marked fluid accumulation and lymphatic tissue damage. It is not unusual for patients to be admitted to the hospital for a course of intravenous antibiotics during an episode of acute cellulitis.41 Infections caused by streptococci are typically treated with beta-lactam antibiotics with activity against bacterial penicillinase. Treatment must be administered promptly to prevent the infection from developing patterns of recurrence. If the infection is caused by methicillin-resistant S. aureus, which is assumed in cases that involve purulent cellulitis without a drainable abscess, vancomycin is the first-line treatment. If vancomycin is not tolerated, fluoroquinolones, trimethoprimsulfamethoxazole, clindamycin, and minocycline may be used.38 For individuals who have three or more episodes of cellulitis per year, long-term prophylactic antibiotic therapy should be considered.42 With the exception of Staphylococcus cellulitis, this usually involves treatment with penicillin V, because most cellulitis is caused by streptococci that are responsive to simple penicillin. For Staphylococcus-positive cellulitis, penicillinase-resistant penicillins are recommended.38 A Cochrane review has investigated the therapeutic benefit of prophylactic antibiotic drugs for reducing the frequency and severity of acute inflammatory episodes in patients with lymphedema. The authors concluded that penicillin can significantly reduce inflammatory episodes if it is combined with proper skin care.29

Drugs for Fibrotic Lymphedema Vitamin E and pentoxifylline have been explored as treatments for fibrosis that arises from iatrogenic lymphedema. Vitamin E (alpha-tocopherol) is a fat-soluble vitamin and a peroxyl radical scavenger that protects lipid membranes from oxidative damage; it also prevents the oxidation of polyunsaturated fatty acids.43 Vitamin E can modulate the expression of connective tissue growth factor, and it is associated with wound repair.44 Pentoxifylline is a methylated xanthine derivate that inhibits cyclic adenosine monophosphate phosphodiesterase to increase blood flow. It also decreases plasma fibrinogen and increases fibrinolytic activity.45 Many studies have reported clinical improvements with these treatments, but only one doubleblind placebo-controlled randomized trial has been conducted.46 Sixty-eight women with unilateral arm edema after breast cancer treatment (axillary/supraclavicular radiotherapy and surgery) who developed a posttreatment increase in arm edema of 20% or more were studied. Thirty-five received oral DL-alpha-tocopherol acetate (500 mg) and pentoxifylline (400 mg) twice daily for 6 months. The volume of the edematous limb was measured with an optoelectronic limb volumeter at 6 and 12 months and reported as a percentage of the volume of the unaffected limb. The degree of fibrosis was also measured. Previous case studies have suggested that combination treatment with vitamin E and pentoxifylline can promote the regression of radiation-induced subcutaneous

K23346_Neligan_30_Pharma Treat_r3_0407-0422.indd 415

5/26/2015 10:00:15 AM

416

Part V  Current Treatment of Lymphedema

fibrosis.47,48 However, Gothard et al46 did not find evidence to support these results in their clinical trial. There was also no indication that this drug combination reduced arm edema.

Caution: Drugs That May Worsen Lymphedema Lymphedema can be understood as an imbalance in fluid transport in which capillary filtration exceeds lymphatic drainage and results in tissue edema.49 In individuals with normal lymphatic function, edema can be induced by drugs that increase capillary filtration beyond the absorptive capacity of the lymphatics. This creates a temporary form of edema that will resolve with cessation of the drug. In patients with lymphedema, the impaired lymphatic system is already overwhelmed by the task of absorbing the body’s basal rate of interstitial fluid. Thus the administration of drugs that further increase capillary filtration will worsen fluid pooling and edema in addition to causing further damage to the lymphatic structures via the dilation of the vessels. This causes incompetent valves, lymph backflow, and reduced lymphatic pumping, all of which contribute to disease progression, more pronounced symptoms, and a poorer quality of life. Drugs can cause increased capillary filtration through three major mechanisms. First, they can promote fluid retention and elevated blood volume; this is typical of corticosteroids, sex hormones, and other hormones. These drugs increase the hydrostatic pressure found in the capillaries. According to the Starling equation, the net movement of fluid across membrane-bound compartments is driven by capillary hydrostatic pressure that exceeds interstitial hydrostatic pressure. The second factor to consider is surface area, because this is also proportional to the rate of diffusion. Drugs that cause vasodilation, such as alpha-blockers and calcium channel blockers, increase the surface area of the peripheral arterioles and allow for more fluid movement into the interstitial spaces. Third, certain drugs increase the interstitial oncotic pressure, which allows pulling of water by osmosis from the capillaries into the interstitial space. The lymphatic vessels in lymphedematous tissues are unable to absorb and recirculate this excess interstitial fluid, and the vascular system is also less likely to reabsorb the fluid as a result of the increased interstitial oncotic pressure. Loop diuretics belong to this category of drugs, and they are considered a paradox in lymphedema management. Although these drugs are responsible for reducing general edema by increasing the renal excretion of water, they are not recommended for patients with pure lymphedema.8 Lymphoscintigraphy has demonstrated no improvement in lymphatic drainage when patients use diuretics, and some patients actually experience reduced lymphatic uptake.10 This may be because loop-diuretic–induced volume depletion contributes to a higher interstitial protein concentration, which can lead to the osmotic exacerbation of high-protein edema in areas of damaged lymphatics. There is also evidence of increased fibrosis in patients with lymphedema who are incorrectly prescribed long-term loop diuretics as a management strategy.50 Granted, there are circumstances in which loop diuretics should be considered (for example, when other fluid-retaining drugs are being taken that are demonstrably exacerbating the patient’s lymphedema). The dosage must be carefully considered, and the drugs should be administered for the shortest duration possible. When patients with lymphedema present after the long-term use of a loop diuretic, discontinuation of the drug should be closely monitored, because rebound edema can occur in patients with hyperaldosteronism caused by long-term diuresis.8

K23346_Neligan_30_Pharma Treat_r3_0407-0422.indd 416

5/26/2015 10:00:15 AM

Chapter 30  Pharmacologic Treatment of Lymphedema

417

Keeley8 systematically searched drug databases such as the Electronic Medicines Compendium and Micromedex for drugs that cause peripheral edema. These drugs are listed and categorized below. Caution is recommended when prescribing these drugs to patients with lymphedema (Box 30-1).

BOX 30-1  Drugs That Cause Peripheral Edema Calcium Antagonists

Antidiabetic Agents

• Amlodipine • Felodipine • Nifedipine

• Pioglitazone • Rosiglitazone (drug has an edema-promoting effect that is increased if it is taken in conjunction with insulin or sulfonylureas)

Corticosteroids • Dexamethasone • Fludrocortisone • Prednisolone

Nonsteroidal Antiinflammatory Drugs • Celecoxib • Diclofenac • Ibuprofen • Naproxen

Alpha-blockers • Doxazosin

Sex Hormones • Anastrozole • Bicalutamide • Estrogen (for example, combined oral contraceptive pills or hormone replacement therapy) • Letrozole • Medroxyprogesterone acetate • Megestrol • Nandrolone • Raloxifene • Tamoxifen • Testosterone • Tibolone

Other Hormones • Paricalcitol • Somatropin

Anticonvulsants • Gabapentin • Pregabalin

Antidepressants • Mirtazapine • Monoamine oxidase inhibitors • Trazodone

K23346_Neligan_30_Pharma Treat_r3_0407-0422.indd 417

Antiparkinsonian Agents • Amantadine • Cabergoline • Ropinirole

Antipsychotics • Fluphenazine • Lithium • Olanzapine • Risperidone

Bisphosphonates • Risedronate • Tiludronate • Zoledronic acid

Cytotoxic Chemotherapy Agent • Docetaxel

Proton Pump Inhibitors • Esomeprazole • Lansoprazole • Omeprazole • Pantoprazole

Other Drugs • Anagrelide • Artorvastatin • Baclofen • Cilostazol • Ciprofloxacin • Etretinate • Glatiramer acetate • Isosorbide dinitrate • Itraconazole • Metoclopramide • Nicotinic acid • Orlistat • Pentoxifylline • Sirolimus • Tacrolimus • Voriconazole

5/26/2015 10:00:15 AM

418

Part V  Current Treatment of Lymphedema

Conclusion Pharmacotherapies are seldom included in best-practice recommendations for the treatment of lymphedema, probably because conservative and surgical options generally result in successful long-term management of the condition. The paucity of methodologically robust clinical trials that verify the efficacy of these drugs may also contribute to their limited recognition. The situation is different for lymphatic filariasis, for which DEC, ivermectin, and doxycycline have been accepted as first-line treatments used to disable resident nematodes and to prevent progressive lymphatic damage and lymphedema. With regard to cellulitis and other acute episodes of inflammation that are common complications of lymphedema, there is sufficient evidence to advocate the use of beta-lactam antibiotics with antipenicillinase properties. Even through the lens of evidence-based medicine, there is still insufficient support for drugs that target high-protein edema; these include lymphedema drugs (benzopyrones), antifibrotic drugs (vitamin E and pentoxifylline), and complementary and alternative pharmacotherapies (herbal remedies and vitamin supplementation with selenium). Although CAM is becoming more widely accepted and sought after, there has been little research into herbal remedies for lymphedema compared with other treatment options. This is an area for future development as herbal therapies demonstrate positive gestalt effects. Herbal mixtures contain many different active ingredients that interact synergistically to produce delayed immune-mediated drug tolerance as well as stronger effects at lower dosages. There are already some exciting results from preliminary studies of traditional herbal mixtures such as saireito51 and juzentaihoto52 that merit larger clinical trials. A review of patent documents also suggests that a number of common herbals, such as cassia cinnamon, hold tremendous potential to improve lymphatic function. As more is understood about lymphatic physiology, drugs with different mechanisms of action for improving lymphedema can also be explored. These mechanisms may include reduction of edema, improvement of lymphatic pumping, and de novo lymphangiogenesis. The fraternal twin of clinical medicine is research, and it must continue to be championed so that health care providers can offer consumers more varied treatment options, including pharmacotherapies. In the meantime, physicians must counsel patients to persevere with their daily tasks of lymphedema management and to be mindful of triggers, such as certain medications, that can exacerbate their symptoms.

K23346_Neligan_30_Pharma Treat_r3_0407-0422.indd 418

5/26/2015 10:00:15 AM

Chapter 30  Pharmacologic Treatment of Lymphedema

419

C linical P earls • With regard to doctor-patient rapport, it is important to acknowledge that adherence to conservative therapies can be difficult. Patient involvement in brainstorming how to overcome their barriers can encourage them to be more active participants in their health care and motivate persistence with their self-care routines. In patients in whom adherence is unachievable, clinicians should empower their patients with information about alternative strategies based on evidence-based medicine. However, patients should be counseled that there are no silver bullet solutions for managing lymphedema and that a multimodal plan is most likely to be effective. • Patients interested in using benzopyrones should be screened for polymorphisms in cytochrome P450 2A6 before the initiation of the pharmacotherapy. This is because inactive or nonfunctional versions of this cytochrome are associated with coumarin-induced hepatotoxicity. • There is little evidence to support the use of HCSE. For patients who insist on a trial of this herbal supplement, the physician should caution them to limit intake to less than 600 mg/day. Patients should be advised that they may have better success with butcher’s broom, sweet clover, or tonka beans if they prefer the herbal option. Alternatively, coumarins and oxerutins may produce superior symptom reduction. • The evidence for selenium is also tenuous, and daily intake should not exceed 400 mg. • Treatment of lymphatic filariasis does not reverse clinical lymphedema. It aims to eliminate the infective organisms before they cause permanent structural damage to the lymphatic system. Thus it should be initiated as quickly as possible. If the individual has concomitant onchocerciasis or is pregnant, it is important to avoid treatment with DEC; ivermectin should be used instead. • Individuals who have had three or more episodes of cellulitis resulting from their lymphedema should be given prophylatic antibiotic therapy—typically penicillin V. • Acute episodes of cellulitis are usually treated with beta-lactams, but vancomycin can be used in methicillinresistant Staphylococcus aureus (MRSA)–positive individuals. • Loop diuretics should be avoided where possible in patients with lymphedema, because this will paradoxically aggravate high-protein edema in areas of damaged lymphatics.

R eferences 1. Földi E, Földi M. Lymphostatic disease. In Földi M, Földi E, eds. Földi’s Textbook of Lymphology for Physicians and Lymphedema Therapists, ed 3. Munich: Urban & Fischer, 2012. 2. Best Practice for the Management of Lymphedema: International Consensus. London: Medical Education Partnership, 2006. 3. Korpan MI, Crevenna R, Fialka-Moser V. Lymphedema: a therapeutic approach in the treatment and rehabilitation of cancer patients. Am J Phys Med Rehabil 90(5 Suppl 1):S69-S75, 2011. 4. Linnitt N. Lymphoedema: recognition, assessment and management. Br J Community Nurs 10:S20S26, 2005. 5. Bogan LK, Powell JM, Dudgeon BJ. Experiences of living with non-cancer-related lymphedema: implications for clinical practice. Qual Health Res 17:213-224, 2007. 6. Planinsek Rucigaj T, Tlaker Zunter V, Miljković J. [Compression therapy for lymphedema: our experience] Acta Med Croatica 64:167-173, 2010.

K23346_Neligan_30_Pharma Treat_r3_0407-0422.indd 419

5/26/2015 10:00:15 AM

420

Part V  Current Treatment of Lymphedema

7. Wang K, Piller N, Conway N, et al. International Lymphedema Framework Australia: emerging issues and the way forward. J Lymphoedema 9:36-40, 2014. 8. Keeley V. Drugs that may exacerbate and those used to treat lymphoedema. J Lymphoedema 3:57-65, 2008. 9. Casley-Smith JR, Casley-Smith JR. Coumarin in the treatment of lymphedema and other high-protein edemas. In O’Kennedy R, Thorne RD, eds. Coumarins: Biology, Applications and Mode of Action. West Sussex: John Wiley & Sons, 1997. 10. Casley-Smith JR, Casley-Smith JR. Drug therapy for lymphedema. In Casley-Smith JR, Casley-Smith JR, eds. Modern Treatment for Lymphedema, ed 5. Malvern: Lymphedema Association of Australia, 1997. 11. Piller NB. A comparison of the effect of benzopyrones and other drugs with anti-inflammatory properties on acid and neutral protease activity levels in various tissues after thermal injury. Br J Exp Pathol 57:411-418, 1976. 12. Badger C, Preston N, Seers K, et al. Benzo-pyrones for reducing and controlling lymphoedema of the limbs. Cochrane Database Syst Rev (2):CD003140, 2004. 13. Twycross R. Drug treatment for lymphedema. In Twycross R, Jenns K, Todds J, eds. Lymphedema. Oxford: Radcliff Medical Press, 2000. 14. Casley-Smith JR. Benzo-pyrones in the treatment to lymphoedema. Int Angiol 18:31-41, 1999. 15. Bensoussan A, Lewith GL. Complementary medicine research in Australia: a strategy for the future. Med J Aust 181:331-333, 2004. 16. MacLennan AH, Wilson DH, Taylor AW. The escalating cost and prevalence of alternative medicine. Prev Med 35:166-173, 2002. 17. Facino RM, Carini M, Stefani R, et al. Anti-elastase and anti-hyaluronidase activities of saponins and sapogenins from Hedera helix, Aesculus hippocastanum, and Ruscus aculeatus: factors contributing to their efficacy in the treatment of venous insufficiency. Arch Pharm (Weinheim) 328:720-724, 2006. 18. Hutson PR, Love RR, Cleary JF, et al. Horse chestnut seed extract for the treatment of arm lymphedema. J Clin Oncol 22(14S):8095, 2004. 19. Huang Y, Kou J, Liu J, et al. Comparison on anti-inflammatory activities of ruscogenin, a major steroidal sapogenin from Radix Ophiopogon japonicus, and its succinylated derivative, RUS-2HS. Drug Dev Res 69:196-202, 2008. 20. Cluzan RV, Alliot F, Ghabboun S, et al. Treatment of secondary lymphedema of the upper limb with CYCLO 3 FORT. Lymphology 29:292-294, 1996. 21. Cluzan RV, Pecking AP, Mathiex-Fortunet H, et al. Efficacy of BN165 (Ginkor Fort) in breast cancer related upper limb lymphedema: a preliminary study. Lymphology 37:47-52, 2004. 22. Pastura G, Mesiti M, Romeo D, et al. [Lymphedema of the upper extremity in patients operated for carcinoma of the breast: clinical experience with coumarinic extract from Melilotus officinalis] Clin Ter 150:403-408, 1999. 23. Vettorello G, Cerreta G, Derwish A, et al. [Contribution of a combination of alpha and beta benzopyrones, flavonoids and natural terpenes in the treatment of lymphedema of the lower limbs at the 2d stage of the surgical classification] Minerva Cardioangiol 44:447-455, 1996. 24. Farinola N, Piller NB. CYP2A6 polymorphisms: is there a role for pharmacogenomics in preventing coumarin-induced hepatotoxicity in lymphedema patients? Pharmacogenomics 8:151-158, 2007. 25. Földi-Börcsök E, Földi M. Lymphedema and vitamins. Am J Clin Nutr 26:185-190, 1973. 26. Sunde RA. Selenium. In Ross AC, Caballero B, Cousins RJ, et al, eds. Modern Nutrition in Health and Disease, ed 11. Philadelphia: Lippincott Williams & Wilkins, 2012. 27. Kasseroller R. Erysipelprophylase beim sekundaren Lymphedemait Selen. Der Allgemeinartzt 3:244247, 1996. 28. Kasseroller R. Sodium selenite as prophylaxis against erysipelas in secondary lymphedema. Anticancer Res 18:2227-2230, 1998.

K23346_Neligan_30_Pharma Treat_r3_0407-0422.indd 420

5/26/2015 10:00:15 AM

Chapter 30  Pharmacologic Treatment of Lymphedema

421

29. Badger C, Seers K, Preston N, et al. Antibiotics/anti-inflammatories for reducing acute inflammatory episodes in lymphoedema of the limbs. Cochrane Database Syst Rev (2):CD003143, 2004. 30. Bruns F, Büntzel J, Mücke R, et al. Selenium in the treatment of head and neck lymphedema. Med Princ Pract 13:185-190, 2004. 31. National Institutes of Health, Office of Dietary Supplements. Selenium: dietary supplement fact sheet, 2013. Available at http://ods.od.nih.gov/factsheets/Selenium-HealthProfessional/. 32. Michael E, Bundy DA, Grenfell BT. Re-assessing the global prevalence and distribution of lymphatic filariasis. Parasitology 112(Pt 4):409-428, 1996. 33. Klion AD. Epidemiology, pathogenesis, and clinical manifestations of lymphatic filariasis. UpToDate 5675(8), 2014. 34. Klion AD. Diagnosis, treatment and prevention of lymphatic filariasis. UpToDate 5680(10), 2014. 35. Critchley J, Addiss D, Gamble C, et al; International Filariasis Review Group. Albendazole for lymphatic filariasis. Cochrane Database Syst Rev (4):CD003753, 2009. 36. Zuther JE. Complications in lymphedema. In Zuther JE, Norton S, eds. Lymphedema Management: The Comprehensive Guide for Practitioners, ed 2. Stuttgart: Theime, 2009. 37. Nowicki J, Siviour A. Best practice skin care management in lymphedema. Wound Pract Res 21:61-65, 2013. 38. McGilvray S. Lymphedema and cellulitis: a narrative review. Wound Pract Res 21:56-60, 2013. 39. Ryan TJ, Mallon EC. Lymphatics and the processing of antigen. Clin Dermatol 13:485-492, 1995. 40. Mallon EC, Ryan TJ. Lymphedema and wound healing. Clin Dermatol 12:89-93, 1994. 41. Jeffs E. The effect of acute inflammatory episodes (cellulitis) on the treatment of lymphoedema. J Tissue Viability 3:51-55, 1993. 42. Mohler ER, Mondry TE. Prevention and treatment of lymphedema. UpToDate 738(19), 2014. 43. Packer J, Fuchs J. Vitamin E in Health and Disease. New York: Dekker, 1993. 44. Villacorta L, Graça-Souza AV, Ricciarelli R, et al. Alpha-tocopherol induces expression of connective tissue growth factor and antagonizes tumor necrosis factor-alpha-mediated downregulation in human smooth muscle cells. Circ Res 92:104-110, 2003. 45. Okunieff P, Augustine E, Hicks JE, et al. Pentoxifylline in the treatment of radiation-induced fibrosis. J Clin Oncol 22:2207-2213, 2004. 46. Gothard L, Cornes P, Earl J, et al. Double-blind placebo controlled randomised trial of vitamin E and pentoxifylline in patients with chronic arm lymphedema and fibrosis after surgery and radiotherapy for breast cancer. Radiother Oncol 73:133-139, 2004. 47. Fischer M, Wohlrab J, Marsch W. Crux medicorum ulcerated radiation-induced fibrosis—successful therapy with pentoxifylline and vitamin E. Eur J Dermatol 11:38-40, 2001. 48. Gottlöber P, Krähn G, Korting HC, et al. [The treatment of cutaneous radiation-induced fibrosis with pentoxifylline and vitamin E. An empirical report] Strahlenther Onkol 172:34-38, 1996. 49. Földi M, Földi E. Physiology and pathophysiology of the lymph vascular system. In Földi M, Földi E, eds. Földi’s Textbook of Lymphology for Physicians and Lymphedema Therapists, ed 3. Munich: Urban & Fischer, 2012. 50. Földi E, Földi M. Lymphostatic diseases. In Földi M, Földi E, Kubik S, eds. Textbook of Lymphology. Munich: Urban & Fischer, 2003. 51. Nagai A, Shibamoto Y, Ogawa K. Therapeutic effects of saireito (chai-ling-tang), a traditional Japanese herbal medicine, on lymphedema caused by radiotherapy: a case series study. Evid Based Complement Alternat Med 2013:241629, 2013. 52. Kogure T, Hoshino A, Ito K, et al. Beneficial effect of complementary alternative medicine on lymphedema with rheumatoid arthritis. Mod Rheumatol 15:445-449, 2005.

K23346_Neligan_30_Pharma Treat_r3_0407-0422.indd 421

5/26/2015 10:00:15 AM

K23346_Neligan_30_Pharma Treat_r3_0407-0422.indd 422

5/26/2015 10:00:15 AM

C hapter 31 Excisional Approaches for the Treatment of Lymphedema Manish C. Champaneria, Peter C. Neligan

Exc K ey P oints • There is still a role for excisional approaches to lymphedema. • Proper patient selection is extremely important. • Although the Charles procedure is rarely performed, it still has a significant role for properly selected patients. • There are numerous variations of the Charles procedure; many of these are very rarely performed and thus are mainly of historical interest. • Liposuction can be considered an excision procedure, and it is extremely useful for the reduction of limb volume (see Chapter 32). • Excisional procedures can be combined with physiologic procedures, such as lymphaticovenular anastomosis and vascularized lymph node transfer.

The surgical treatment of lymphedema has advanced with the introduction of microsurgical lymph node transfer and lymphatic venous bypass. However, countless patients still have debilitating lymphedema that is not amenable to such sophisticated procedures because of the surgeon’s lack of appropriate expertise, the patient’s disease status, or patient or surgeon preference. Excisional surgeries are sometimes radical, but they are often the only treatment modalities appropriate for these patients. Such patients are frequently considered to have inoperable advanced disease, and they become victims of medical systems that refer them from physician to physician while offering dwindling treatment options. Most early methods for the treatment of lymphedema, such as reductive management and radical excision, have been abandoned over the past century. However, the principles of such surgeries remain pertinent to patients with end-stage lymphedema. For carefully selected patients, these techniques can offer a significant amount of satisfaction and excellent end results. 423

K23349_Neligan_31_Excisional_r5_dc_0423-0436.indd 423

5/27/2015 9:36:42 AM

424

Part V  Current Treatment of Lymphedema

As discussed throughout this book, lymphedema is a chronic pathologic state of the lymphatic system. Lymph accumulates in the subcutaneous tissues, where it causes increased volume and weight of the limb, skin tension, and disfigurement.1 Lymphedema is generally categorized as congenital, infectious, or iatrogenic, although its exact cause is still not fully understood. In the United States, lymphedema occurs most commonly as a sequela of the treatment of a malignancy.2-6 Elsewhere, especially in parts of the developing world, infection with Wuchereria bancrofti (filariasis) is the most common cause. Lymphedema has traditionally been viewed as incurable or intractable. However, during recent years there have been significant advances in lymphedema genetics, diagnostic modalities, and conservative treatments (see Chapters 8 and 12). Nonsurgical treatments such as complex decongestive therapy and compressive garments are considered palliative; the aim is to prevent disease progression and provide symptomatic relief. These treatments are expensive and time consuming, have poor compliance rates, and do not address the changes that arise in patients with severe endstage disease with chronic fibrosis.7 Surgical reduction is a viable treatment option for patients with disfiguring and advanced lymphedema. In this chapter we describe both earlier and current excisional surgical treatment options for advanced lymphedema.

Patient Selection The management of lymphedema has traditionally focused on conservative modalities. These medical treatments—compression therapy, complex decongestive physiotherapy using lymphedemaspecific manual lymphatic drainage massage, and external sequential pneumatic compression devices—are often attempted before surgical therapy is considered.8 These conservative modalities to enhance drainage have demonstrated success in cases of mild to moderate lymphedema, but they are associated with high patient-to-patient variability, partly because their success depends on patient compliance.9 Discouragingly, a significant number of patients receive little benefit. In addition, these methods can be costly, time consuming, and uncomfortable, which reduces patient compliance still further.7,10 The basis of this conservative approach stems partly from the idea that surgical intervention leads to considerable morbidity. Because surgery for lymphedema has a high complication rate that includes recurrence, infection, and wound-healing problems, it is often delayed.11 Many clinicians recommend at least 2 years of conservative therapy before any form of surgical intervention is considered.12 For patients with end-stage disease and large, fibrotic limbs, conservative therapy is often ineffective or is not properly instituted. For these patients excisional surgery has a definitive role and must be considered. Lymphedema surgery involves reductive and reconstructive procedures. Reconstructive techniques rebuild the lymphatic channels through vascularized lymph node transfers, anastomoses between lymphatic channels and veins, and lymphatic grafts using lymphatic-lymphatic anas­ tomoses. These surgeries are beneficial during the early stages of lymphedema; however, they have not resulted in consistent improvement in patients with advanced disease because of the relatively minimal reduction in the fibrosclerotic tissue that is seen after lymphatic reconstruction is per-

K23349_Neligan_31_Excisional_r5_dc_0423-0436.indd 424

5/27/2015 9:36:43 AM

Chapter 31  Excisional Approaches for the Treatment of Lymphedema

425

formed.13 The role of microsurgery is generally limited to early disease, when the lymphatics are relatively healthy and the tissues are still soft and pliable. Chronically accumulated lymphatic fluid in the subcutaneous tissues causes cutaneous thickening, hypercellularity, progressive fibrosis, and increased adipose tissue.14 Damage to lymphatic vessels is irreversible after prolonged disease, and attempts to restore lymph flow will be inadequate if they are attempted in patients with end-stage disease. Patients with advanced International Society of Lymphology stage III disease— elephantiasis with severe limb deformation, scleroindurative pachydermatitis, and widespread lymphostatic warts—are the ideal candidates for excisional surgical procedures.15 Sometimes excisional and reconstructive techniques can be combined to enhance patient outcomes and improve aesthetics.

Excisional Techniques The Charles Procedure Reductive or excisional therapy is appropriate when conservative measures fail, when the extremities become massive and fibrotic, and when the disease is incapacitating. The surgical technique for this type of treatment is typified by the Charles procedure, which was named for Sir Richard Henry Havelock Charles and first described by him in 1912.16 During the mid-1880s, Charles served as the chief medical officer of the Afghan Boundary Commission, which surveyed the land between British India and the Russian Empire. His service ultimately brought him to Calcutta, where he became a professor of surgery and anatomy at Bengal Medical College.17 It is here that he first documented his technique of applying split-thickness skin grafts to debulked tissue for the treatment of lymphedema.16 Although Charles did describe excisional surgery for advanced filarial lymphedema, his technique pertained primarily to the scrotum. He documented one case of lower extremity lymphedema that was treated in this manner, but it was unsuccessful.18 Despite a paucity of information about Charles’s treatment method, the procedure bears his name as a result of a series of references that can be traced back to a questionable citation by the British plastic surgeon Sir Archibald McIndoe.18,19 The Charles procedure involves total subcutaneous excision that removes virtually all skin, subcutaneous tissue (except in the foot and the region overlying the calcaneal tendon), and deep fascia; the bare muscle is subsequently reconstructed using skin grafts. Because there is no subdermal plexus for drainage and the skin graft is adherent to bare muscle, much worse edema is produced distal to the excision, usually in the foot. Severe secondary skin changes—including ulceration, papillomatosis, hyperkeratosis, keloid formation, hyperpigmentation, and a weeping dermatitis—in addition to chronic cellulitis frequently occur, especially if split-thickness grafts are used.20 The extremity should ideally be resurfaced with more durable full-thickness grafts to prevent secondary deformity, although harvesting full-thickness grafts of a sufficient quantity is a challenge.21 Most surgeons have modified the original technique to preserve the deep fascia; they then graft onto the fascia rather than the bare muscle. When this procedure is performed correctly for the carefully selected patient, it safely provides a consistent reduction in size, an improvement in function, and satisfactory results, although it may be deemed aesthetically unsatisfactory. Because of its poor cosmesis and associated bottleneck deformity, this procedure has almost been abandoned (Fig. 31-1).

K23349_Neligan_31_Excisional_r5_dc_0423-0436.indd 425

5/27/2015 9:36:43 AM

426

Part V  Current Treatment of Lymphedema

A

B

C

FIG. 31-1  The Charles procedure. A, A patient with a lymphedematous lower extremity. The patient had a history of filariasis and had been offered an amputation. B, The Charles procedure involves the complete circumferential excision of the skin and subcutaneous tissue of the involved leg. The dorsum of the foot had previously been excised and grafted. C, Reconstruction of the lower extremity is performed with either split-thickness skin grafting or full-thickness skin grafting using the excised skin. The patient is then evaluated 6 months after surgery.

Variations of the Charles Procedure As the Charles procedure became more popular and Charles’s name recognition grew, several variations of the procedure were described. Sistrunk, Auchincloss, Homans, Macey, and Thompson all contributed to variations of the Charles procedure.22-26 During these procedures, the limb was generally significantly reduced by excising an ellipse of the skin and soft tissue of the extremity and creating dermal flaps to avoid excessive skin grafting; variations arose with regard to the management of the underlying fascia and the method used to foster the drainage of superficial lymph channels through the deep collecting system. The following list presents a brief summary of the variations of the Charles procedure, which are described here for completeness and acknowledgment in surgical history: • The Kondoleon procedure (1912): The Kondoleon procedure was one of the earliest variations, and it involved the excision of the lymphedematous tissue as well as vertical strips of deep fascia; this was done to open the deep lymphatics to create communication between the superficial and deep lymphatics. Kondoleon believed that the fascia covering the muscles separated the superficial and deep lymphatic systems, and the goal of his procedure was to connect these two systems by removing the fascia. A large amount of skin and subcutane-

K23349_Neligan_31_Excisional_r5_dc_0423-0436.indd 426

5/27/2015 9:36:43 AM

Chapter 31  Excisional Approaches for the Treatment of Lymphedema

427

ous fat would be excised, and the skin was then allowed to become attached to the muscles. When the raw surfaces were opposed, it was believed that new blood vessels, nerves, and lymphatics formed; thus the superficial lymphatic system would be connected with the deep system. However, the creation of a fascial window apparently did not work well. Only the tissue resection part of this procedure continues to be used and to be erroneously referred to as the Kondoleon procedure. • The Sistrunk procedure (1918): The Sistrunk procedure was a modification of the Kondoleon procedure. Walter Ellis Sistrunk (1880-1933) was an outstanding surgeon who spent a significant amount of his career as an associate professor of surgery at the Mayo Clinic in Rochester, Minnesota. He is most known for an operation that cured thyroglossal duct cysts, and he excelled at the surgical treatment of the thyroid, breast, and colon. The lymphedematous tissue was excised, and larger window cuts in the deep fascia were created to allow for communication into the normal deep lymphatics. Sistrunk believed that the removal of large amounts of fascia was one of the very important modifications of his procedure. After the wedge excision of the skin and the lymphedematous tissue, the wound was sutured together for primary closure. This procedure is still commonly used when a large lymphedematous pannus requires excision, but the fascia is often left intact. • The Homans-Miller procedure (1936): The Homans-Miller procedure was a modification of the Kondoleon procedure that involved the use of particularly thin skin flaps to cover the resected area. With these flaps, Miller was able to achieve an aesthetically pleasing result. He elevated anterior and posterior flaps from both medial and lateral incisions; the flaps were approximately 1 cm thick. The underlying lymphedematous tissue was excised down to the muscle fascia, and the skin flaps were trimmed and sutured into position. Good aesthetic and functional results were obtained with this procedure, which is now considered by some to be the standard ablative approach for the treatment of forearm and upper extremity lymphedema. However, occasionally second or even third operations are required to obtain the maximum benefit. • The Thompson procedure (1962)26: The Thompson procedure was a combination type that incorporated techniques from both the Charles procedure and the Homans-Miller procedure. It uses the deepithelialized segment of skin as a kind of wick that was thought to absorb fluid, as well as to serve as a communication between the superficial and deep lymphatic systems. However, this is no longer used. Lymphedematous tissue under the skin flaps was excised, and the epidermis and part of the dermis of one of the skin flaps were shaved off with a Humby knife. This shaved flap was then buried under the opposite flap, deep to the deep fascia, like a Swiss roll. • The Macey procedure22: This is a variation of the other procedures and is no longer used. The skin and subcutaneous tissue were peeled back with the deep fascia, and split-thickness skin grafting was performed over the denuded area. The overlying pad of tissue was sutured back temporarily and then trimmed away 10 days later. Kondoleon added the excision of strips of deep fascia to Charles’s procedure. Sistrunk modified Kondoleon’s procedure to excise greater amounts of fascia and subcutaneous tissue, and he introduced the raising of a skin flap to close the wound primarily. However, others reported that the deep fascia regrew within 3 months and that the uptake of lymph was the result of the rich vascular bed of the muscle rather than its lymphatic system.27 When both the deep and superficial lymphatic systems were excised, the rich, vascular muscle bed remained, as proposed, the mechanism for lymph dispersal.28 In 1962 Thompson29 modified these procedures again with the belief

K23349_Neligan_31_Excisional_r5_dc_0423-0436.indd 427

5/27/2015 9:36:43 AM

428

Part V  Current Treatment of Lymphedema

that dermis, with its rich lymph supply, drained directly into the muscle rather than depending on transmission through the subcutaneous tissue. Harvey30 described improved lymph flow as measured by radioiodinated human serum albumin after the Thompson procedure; others failed to corroborate this finding entirely and believed the improvements were the result of the excision of subcutaneous tissue. When cutaneous nerves were preserved and the excisional surgeries staged, as described by Miller et al,31 good results were observed in clinical studies; improved radioiodinated human serum albumin clearance was also noted. Despite the many permutations of the excisional surgery, preoperative and postoperative care remained constant: strict bed rest to decrease edema. Miller et al31 even suggested suspending the leg from an overhead bed frame with a Thomas splint to provide dependent drainage.  These approaches provided the most reasonable surgical compromise by offering reliable improvement and a minimum of unfavorable postoperative complications. Improvement was directly related to the amount of skin and subcutaneous tissue removed as well as to the personal care and attitude of each patient. These procedures all allowed improved skin hygiene, decreased risk of cellulitis and sepsis, and the easier application of compression garments.32 However, the Charles procedure and its variations have been largely forgotten because of the availability of nonoperative treatments, the potential morbidity of such radical surgeries, and the associated complications. These ideas are rooted in many of the original descriptions of excisional surgery that date back to the early 1900s, when aggressive debulking was used. At that time, difficulty maintaining a clean operative field when addressing large wounds may have negatively affected results.18 Furthermore, these procedures were applied to all patients with lymphedema, without specific selection criteria and regardless of the cause, stage, or type of lymphedema present.33 These excisional surgeries were once appreciated and used widely. However, since the middle of the last century, there have been few reported cases in the literature. A recent review showed that during the past 50 years, only 147 patients who had undergone the Charles procedure or one of its modifications were reported in the literature. The earlier cases demonstrated poor results and substantial complications, including infections, poor wound healing, prolonged hospitalizations, long surgical scars, sensory nerve loss, graft necrosis, scar contracture, and the exacerbation of lymphedema below the excised area.21,34 All of these complications arose from one major source of morbidity: difficulties encountered when handling large skin grafts.35 These problems were amplified when the area to be grafted was extensive, such as a lower extremity. A total of six contemporary articles (excluding case reports) were identified as involving excisional procedures for the surgical treatment of lymphedema11,32,36-39 (Table 31-1). Only two of the studies, both by Salgado et al, reported outcomes of volume reduction as a percentage reduction.37,38 The mean follow-up time of these studies ranged from 13 months to 72 months, and variability with regard to study design, lymphedema staging, patient selection, and measurement technique compounded the results of these surgeries when they were analyzed as a group. Again, the paucity of data largely reflects the view of many that these procedures cause significant morbidity. Recent developments and technologic innovations have repopularized excisional surgeries by allowing for minimal complications and good functional outcomes. Karri et al32 reported subjective improvements in mobility with acceptable cosmetic results and low complication rates after performing the Charles procedure on 27 patients for the treatment of chronic lower extremity lymphedema. Specific variations in technique were made to reduce complications, including excision of lymphedematous tissue over the dorsum of the foot and the use of long skin strips to

K23349_Neligan_31_Excisional_r5_dc_0423-0436.indd 428

5/27/2015 9:36:43 AM

429

Chapter 31  Excisional Approaches for the Treatment of Lymphedema

TABLE 31-1  Published Studies of Excisional Procedures for Lymphedema

Study Design

Number of Patients

Modolin et al (2006)39

Prospective

Salgado et al (2007)37

Author (year)

Follow-up (months)

Volume Reduction (%)

Measurement Technique Not reported

Site

Procedure

17

Penis/ scrotum

Excision

72

Not reported

Retrospective

15

Lower extremity

Excision

13

66

Circumference

Lee et al (2008)11

Retrospective

22

Lower extremity

Excision

48

Not reported

Volumetry and circumference

Salgado et al (2009)38

Prospective

11

Upper extremity

Excision

18

21

Circumference

van der Walt et al (2009)36

Retrospective

8

Lower extremity

Excision with negative pressure wound therapy

27

Not reported

Not reported

Karri et al (2011)32

Retrospective

27

Lower extremity

Excision

22

Not reported

Not reported

reduce hypertrophic scarring. van der Walt et al36 demonstrated a modification of the Charles procedure by adding negative pressure wound therapy to the wound bed directly after excisional surgery to improve the bed’s quality for grafting. The results were uniformly good among the eight patients described, with no major complications and significant improvements in assessed functional scores. Lee et al11 reported the successful treatment of end-stage lymphedema with a modified Auchincloss-Homan operation. The authors reported a low complication rate and better overall volume reduction as compared with the control group, which received conservative therapy. The importance of continued compression therapy postoperatively to maintain initial results was stressed. The Charles procedure has also been combined with vascularized lymph node transfers. Sapountzis et al40 performed the first combined radical excisional procedure with vascularized lymph node transfer to reduce the need for secondary operations as well as the risk of recurrence. The use of negative pressure wound therapy greatly simplifies the management of large skin grafts, so it is our current practice to occasionally offer the Charles procedure to selected patients with extreme disease.

Staged Subcutaneous Excision Beneath Skin Flaps Staged subcutaneous excision beneath skin flaps is described here in detail as an example of the excisional techniques used today. This procedure has demonstrated long-lasting results and is commonly used by practices in the United States and around the world.31 Thirty-eight descriptions of this technique have been found in the literature, with long-term follow-up of 3 to 17 years. Substantial improvement, defined as a significant reduction in extremity size, improved functional recovery, and decreased incidence or absence of cellulitis, was found in at least 75% of patients. Before surgery, absolute bed rest and extremity elevation are mandated for 3 days to reduce edema and increase skin laxity to facilitate greater amounts of excision. Bed rest and elevation

K23349_Neligan_31_Excisional_r5_dc_0423-0436.indd 429

5/27/2015 9:36:44 AM

430

Part V  Current Treatment of Lymphedema

A MEDIAL RESECTION

Extent and depth of dissection Incision Lymphedematous tissue

Extent of dissection

Fascia Incision

Sural nerve

Sural nerve Medial malleolus

Redundant skin

B LATERAL RESECTION (3 months postoperatively)

Extent and depth of dissection

Incision Peroneal nerve

Incision Fascia

Extent of dissection Peroneal nerve Lateral malleolus

Redundant skin

FIG. 31-2  Staged subcutaneous excision beneath skin flaps. A, Cross-section (right, top) of a limb showing the incision lines and planned deepithelialized segment. Proposed excision (right, middle) of the lymphedematous tissue. The thin skin flaps are then sutured together and given an aesthetically satisfactory closure. B, The procedure can be repeated 3 months later with a lateral incision as shown, completing the same steps.

K23349_Neligan_31_Excisional_r5_dc_0423-0436.indd 430

5/27/2015 9:36:44 AM

Chapter 31  Excisional Approaches for the Treatment of Lymphedema

431

are performed at home, and the patient is admitted 24 hours preoperatively. Sequential external pneumatic compression, bladder catheterization, and perioperative antibiotics are administered. The surgery is performed in two stages. A medial resection is performed first, because more tissue can be removed from the medial aspect than the lateral aspect of both the arm and the leg. The lateral resection, if needed, is performed 3 months later (Fig. 31-2). If bilateral disease is present, the operation may be performed on both involved limbs during the initial procedure. However, for patients with massive edema, the prolonged operative time and excessive blood loss lessen the appeal of this approach. The leg is exsanguinated, and a tourniquet is used proximally on the extremity. A medial incision is made 1 cm posterior to the medial malleolus and extended proximally into the thigh. Anteriorly and posteriorly based flaps that are 1.5 cm thick are elevated to the midsagittal plane in the calf, with less extensive dissection in the thigh and ankle. Subcutaneous tissue beneath the flaps is removed. The sural nerve is protected, and the fascia around the knee is maintained. Redundant skin is resected, the wound is closed in a single cutaneous layer, and drains are placed. The extremity is immobilized in a posterior splint, and the patient is kept on bed rest, with the extremity elevated. The drains are removed 5 days after surgery; the sutures are removed 9 days after surgery, and tape reinforcement is placed. The extremity is fitted for compression garments, which are used continuously for 3 weeks to prevent seroma formation and wound breakdown. The lateral surgery can be performed 3 months later using a similar technique. Excisional techniques can also be performed with other reconstructive procedures. The excision of difficult-to-manage folds and excesses while performing reconstructive procedures on the remaining lymphatic system has a definite place in the treatment of lymphedema and can lead to improved outcomes (Figs. 31-3 and 31-4).

A

B

FIG. 31-3  A, A patient with stage III lymphedema with a large fold of lymphedematous tissue on her left proximal thigh. B, The same patient is shown after excision surgery. Note the lymphedematous abdominal pannus, which was subsequently excised. Preoperative MRI indicated functioning lymphatics in the distal lower extremity, so this patient was also successfully treated with lymphaticovenular anastomosis.

K23349_Neligan_31_Excisional_r5_dc_0423-0436.indd 431

5/27/2015 9:36:45 AM

432

Part V  Current Treatment of Lymphedema

A

B

C

FIG. 31-4  A, This patient developed left upper extremity lymphedema after mastectomy, axillary node dissection, and radiotherapy. B, Planned brachioplasty at the time of axillary scar excision and vascularized lymph node transfer. C, Early postoperative appearance.

Liposuction Newly developed excisional techniques involving the removal of lymphedematous adipose tissue through suction-assisted lipectomy (liposuction) have proved successful for many patients with lymphedema, including women with upper extremity lymphedema after breast cancer therapy as well as patients with lower extremity lymphedema.41-43 Brorson et al41,42,44,45 described the results of prospectively followed patients and reported favorable results overall. Liposuction has also been combined with the excision of excess skin and major reduction surgery as a neoadjuvant technique to facilitate wound closure, to reduce wound healing problems, and to prevent relapse. Only a few case reports of this technique have been cited in the literature, but similar cases of combined liposuction and excess skin and subcutaneous tissue excision have been described in the body-contouring literature.46 In general, circumferential liposuction for lymphedema is safe; few postoperative complications have been reported, including paresthesias and minor wound healing issues. Cadaver and imaging studies have demonstrated that liposuction does not disrupt lymphatic vessels (if performed parallel to the limb) and that the treatment of lymphedema with liposuction does not decrease the already impaired lymphatic transport capacity of the limb.47-49 The minimal recurrence and complication rates of these recent case series

K23349_Neligan_31_Excisional_r5_dc_0423-0436.indd 432

5/27/2015 9:36:45 AM

Chapter 31  Excisional Approaches for the Treatment of Lymphedema

433

demonstrate how the consideration of disease severity and the refinement of the technique in the modern era of surgical care can support the use of excisional therapy. Liposuction is described in greater detail in Chapter 32.

Future Directions Lymphedema is a complex clinical entity with myriad causes that at present has no cure. Patients have typically suffered from the condition for many years; they often have seen multiple specialists and tried various treatments before presenting to the surgeon with worsening signs and symptoms. With more than 140 million people worldwide suffering from lymphedema, patients with advanced disease will be interested in access to specialized lymphedema clinics. Such tertiary care clinics offer focused health care practitioners, an enhanced surgical experience, and a better understanding of the pathophysiology of lymphedema. The history of excisional surgery for lymphedema contains several sound procedures that may have fallen out of favor as a result of their radical nature and the lack of familiarity that today’s surgeons have with the disease process itself. For example, the Charles procedure—and other excisional procedures based on it—is an extremely challenging operation that poses a risk for severe complications, some of which have been described in the literature. An important component of determining whether the excisional surgical treatment of lymphedema is an option is assessing the risk-benefit ratio of a specific procedure for a specific patient. The individual goals of the patient (for example, functional versus cosmetic), the extent of the surgical procedure, and the level of expertise and experience required to perform the surgery should also be carefully considered. Another critical step when performing excisional surgery for lymphedema is improved patient evaluation and follow-up. Additional studies—prospective or controlled studies as well as studies that compare different treatments—are required to select appropriate patient populations that would derive the greatest benefit from surgery. Although the surgical principles of the excisional treatment of lymphedema are simple and hardly new, the tackling of these problems and the care of these patients may benefit from continued diligence.

C linical P earls • Excision of redundant tissue should be considered, even if contemplating a physiologic procedure. • The Charles procedure still has a place in a small number of selected patients. • Liposuction is a very useful tool in the treatment of lymphedema.

K23349_Neligan_31_Excisional_r5_dc_0423-0436.indd 433

5/27/2015 9:36:45 AM

434

Part V  Current Treatment of Lymphedema

R EFERENCES 1. Rockson SG, Miller LT, Senie R, et al. American Cancer Society Lymphedema Workshop. Workgroup III: Diagnosis and management of lymphedema. Cancer 83:2882-2885, 1998. 2. Ahmed RL, Prizment A, Lazovich D, et al. Lymphedema and quality of life in breast cancer survivors: the Iowa Women’s Health Study. J Clin Oncol 26:5689-5696, 2008. 3. Beaulac SM, McNair LA, Scott TE, et al. Lymphedema and quality of life in survivors of early-stage breast cancer. Arch Surg 137:1253-1257, 2002. 4. Franks PJ, Moffatt CJ, Doherty DC, et al. Assessment of health-related quality of life in patients with lymphedema of the lower limb. Wound Repair Regen 14:110-118, 2006. 5. Ganz PA. The quality of life after breast cancer—solving the problem of lymphedema. N Engl J Med 340:383-385, 1999. 6. Pyszel A, Malyszczak K, Pyszel K, et al. Disability, psychological distress and quality of life in breast cancer survivors with arm lymphedema. Lymphology 39:185-192, 2006. 7. Shih YC, Xu Y, Cormier JN, et al. Incidence, treatment costs, and complications of lymphedema after breast cancer among women of working age: a 2-year follow-up study. J Clin Oncol 27:2007-2014, 2009. 8. Tiwari A, Cheng KS, Button M, et al. Differential diagnosis, investigation, and current treatment of lower limb lymphedema. Arch Surg 138:152-161, 2003. 9. Ko DS, Lerner R, Klose G, et al. Effective treatment of lymphedema of the extremities. Arch Surg 133:452-458, 1998. 10. Mehrara BJ, Zampell JC, Suami H, et al. Surgical management of lymphedema: past, present, and future. Lymphat Res Biol 9:159-167, 2011. 11. Lee BB, Kim YW, Kim DI, et al. Supplemental surgical treatment to end stage (stage IV-V) of chronic lymphedema. Int Angiol 27:389-395, 2008. 12. Lee BB. Contemporary issues in management of chronic lymphedema: personal reflection on an experience with 1065 patients. Lymphology 38:28-31, 2005. 13. Campisi C, Bellini C, Campisi C, et al. Microsurgery for lymphedema: clinical research and long-term results. Microsurgery 30:256-260, 2010. 14. Jensen MR, Simonsen L, Karlsmark T, et al. Lymphoedema of the lower extremities—background, pathophysiology and diagnostic considerations. Clin Physiol Funct Imaging 30:389-398, 2010. 15. International Society of Lymphology. The diagnosis and treatment of peripheral lymphedema: 2013 Consensus Document of the International Society of Lymphology. Lymphology 46:1-11, 2013. 16. Charles RH. Elephantiasis scroti. In Latham AC, English TC, eds. A System of Treatment. London: Churchill, 1912. 17. Sir Havelock Charles, Br, GCVO, LLD, MD. Br Med J 2:838-839, 1934. 18. Dumanian GA, Futrell JW. The Charles procedure: misquoted and misunderstood since 1950. Plast Reconstr Surg 98:1258-1263, 1996. 19. McIndoe A. Treatment of chronic oedema of the leg. Proc R Soc Med 43:1043-1045, 1950. 20. Taylor GW. Surgical management of primary lymphoedema. Proc R Soc Med 58:1024-1026, 1965. 21. Miller TA. Charles procedure for lymphedema: a warning. Am J Surg 139:290-292, 1980. 22. Macey HB. A surgical procedure for lymphoedema of the extremities; a follow-up report. J Bone Joint Surg Am 30:339-346, 1948. 23. Auchincloss H. New operation for elephantitis. PR J Public Trop Med 6:149-150, 1930. 24. Homans J. The treatment of elephantiasis of the legs, preliminary report. N Engl J Med 215:1099, 1936. 25. Sistrunk WE. Further experiences with the Kondoleon operation for elephantiasis. JAMA 71:800-806, 1918. 26. Thompson N. The surgical treatment of chronic lymphoedema of the extremities. Surg Clin North Am 47:445-503, 1967. 27. Berthwhistle AP, Gregg AL. Elephantiasis. Br J Surg 16:267, 1928.

K23349_Neligan_31_Excisional_r5_dc_0423-0436.indd 434

5/27/2015 9:36:45 AM

Chapter 31  Excisional Approaches for the Treatment of Lymphedema

435

28. Peer LA, Shahgholi M, Walker JC Jr, et al. Modified operation for lymphedema of the leg and arm. Plast Reconstr Surg (1946) 14:347-350, 1954. 29. Thompson N. Surgical treatment of chronic lymphoedema of the lower limb. With preliminary report of new operation. Br Med J 2:1566-1573, 1962. 30. Harvey RF. The use of 131I labelled human serum albumin in the assessment of improved lymph flow following buried dermis flap operation in cases of post-mastectomy lymphoedema of the arm. Br J Radiol 42:260-265, 1969. 31. Miller TA, Wyatt LE, Rudkin GH. Staged skin and subcutaneous excision for lymphedema: a favorable report of long-term results. Plast Reconstr Surg 102:1486-1498; discussion 1499-1501, 1998. 32. Karri V, Yang MC, Lee IJ, et al. Optimizing outcome of Charles procedure for chronic lower extremity lymphoedema. Ann Plast Surg 66:393-402, 2011. 33. Lee BB, Laredo J, Neville R. Contemporary indications and controversies. In Lee BB, Bergan JJ, Rockson SG, eds. Lymphedema: A Concise Compendium of Theory and Practice. London: Springer, 2011. 34. Savage RC. The surgical management of lymphedema. Surg Gynecol Obstet 160:283-290, 1985. 35. Gloviczki P. Principles of surgical treatment of chronic lymphoedema. Int Angiol 18:42-46, 1999. 36. van der Walt JC, Perks TJ, Zeeman BJ, et al. Modified Charles procedure using negative pressure dressings for primary lymphedema: a functional assessment. Ann Plast Surg 62:669-675, 2009. 37. Salgado CJ, Mardini S, Spanio S, et al. Radical reduction of lymphedema with preservation of perforators. Ann Plast Surg 59:173-179, 2007. 38. Salgado CJ, Sassu P, Gharb BB, et al. Radical reduction of upper extremity lymphedema with preservation of perforators. Ann Plast Surg 63:302-306, 2009. 39. Modolin M, Mitre AI, da Silva JC, et al. Surgical treatment of lymphedema of the penis and scrotum. Clinics (Sao Paulo) 61:289-294, 2006. 40. Sapountzis S, Ciudad P, Lim SY, et al. Modified Charles procedure and lymph node flap transfer for advanced lower extremity lymphedema. Microsurgery 34:439-447, 2014. 41. Brorson H. Liposuction in arm lymphedema treatment. Scand J Surg 92:287-295, 2003. 42. Brorson H, Svensson H. Liposuction combined with controlled compression therapy reduces arm lymphedema more effectively than controlled compression therapy alone. Plast Reconstr Surg 102:10581067; discussion 1068, 1998. 43. Greene AK, Slavin SA, Borud L. Treatment of lower extremity lymphedema with suction-assisted lipectomy. Plast Reconstr Surg 118:118e-121e, 2006. 44. Brorson H. Liposuction gives complete reduction of chronic large arm lymphedema after breast cancer. Acta Oncol 39:407-420, 2000. 45. Brorson H, Ohlin K, Olsson G, et al. Controlled compression and liposuction treatment for lower extremity lymphedema. Lymphology 41:52-63, 2008. 46. Wollina U, Heinig B, Schönlebe J, et al. Debulking surgery for elephantiasis nostras with large ectatic podoplanin-negative lymphatic vessels in patients with lipo-lymphedema. Eplasty 14:e11, 2014. 47. Brorson H, Svensson H, Norrgren K, et al. Liposuction reduces arm lymphedema without significantly altering the already impaired lymph transport. Lymphology 31:156-172, 1998. 48. Frick A, Hoffmann JN, Baumeister RG, et al. Liposuction technique and lymphatic lesions in lower legs: anatomic study to reduce risks. Plast Reconstr Surg 103:1868-1873; discussion 1874-1875, 1999. 49. Hoffmann JN, Fertmann JP, Baumeister RG, et al. Tumescent and dry liposuction of lower extremities: differences in lymph vessel injury. Plast Reconstr Surg 113:718-724; discussion 725-726, 2004.

K23349_Neligan_31_Excisional_r5_dc_0423-0436.indd 435

5/27/2015 9:36:45 AM

K23349_Neligan_31_Excisional_r5_dc_0423-0436.indd 436

5/27/2015 9:36:45 AM

C hapter 32 Liposuction Håkan Brorson

K ey P oints • Liposuction for lymphedema is indicated when pitting is absent or minimal. • If significant pitting (greater than 8 to10 mm) is present, conservative treatment (combined physical therapy) is indicated to remove the lymph and transfer the lymphedema to a nonpitting state. • This nonpitting state indicates that any remaining excess volume consists of lymphedema-induced adipose tissue.

Lip

• In these patients, further reduction of the excess volume is not possible with conservative treatment or microsurgical reconstructions; only liposuction can remove this excess volume. • As with conservative treatment, the lifelong 24-hour-a-day use of compression garments is essential to maintain the effects of treatment. • Excess volume without pitting means that the adipose tissue is responsible for the swelling.

Some controversy surrounds the use of liposuction to treat late-stage lymphedemas. Although conservative therapies, such as complex decongestive therapy and controlled compression therapy, should clearly be used initially, the treatment options for late-stage lymphedema that does not respond to therapy are not so clear. Moreover, various microsurgical procedures are often performed without the knowledge of adipose tissue deposition, which cannot be removed with these techniques. In contrast to microsurgical procedures, liposuction enables the complete removal of the deposited adipose tissue, leading to complete reduction in late-stage lymphedemas.

Chronic Lymphedema Leading to Adipose Tissue Deposition Several possible explanations for adipose tissue hypertrophy in lymphedema have been postulated. One possibility is the physiologic imbalance of blood flow and lymphatic drainage, which results in the impaired clearance of lipids and their uptake by macrophages.1-3 However, support 437

K23346_Neligan_32_Liposuction_r4_dc_0437-0446.indd 437

5/26/2015 10:01:41 AM

438

Part V  Current Treatment of Lymphedema

is increasing for the idea that the fat cell is an endocrine organ and a cytokine-activated cell4,5 and that chronic inflammation plays a role.1,6,7 Harvey et al8 and Schneider et al9 discussed the relationship between slow lymph flow and adiposity and structural changes in the lymphatic system and adiposity. Other indications for adipose tissue hypertrophy include the following1: • The discovery of increased adipose tissue in intestinal segments in patients with inflammatory bowel disease (Crohn’s disease), known as fat wrapping, has clearly demonstrated that inflammation has a key role.6,10,11 • Consecutive analyses of the content of the aspirate that was removed under bloodless conditions with a tourniquet had a high content of adipose tissue (mean 90%).12 • In Graves’ ophthalmopathy with exophthalmos, adipocyte-related immediate early genes are overexpressed, and cysteine-rich, angiogenic inducer 61 may have a role in both orbital inflammation and adipogenesis.13 • Tonometry can distinguish if a lymphedematous arm is harder or softer than the normal arm. Patients who have a harder arm compared with the healthy arm have excess adipose tissue.14 • Volume-rendered computer tomography in eight patients also showed a significant preoperative increase of 81% in adipose tissue in the swollen arm. This was followed by normalization at 3 months, which paralleled the complete reduction of the excess volume.15,16 • Analyses with dual-energy x-ray absorptiometry in 18 women with postmastectomy arm lymphedema showed a significant increase in adipose tissue in the nonpitting swollen arm before surgery. Postoperative analyses showed normalization at 3 months. This effect was also seen at 12 months. These results paralleled the complete reduction of the excess volume (edema volume).17 • A functional inactivation of a single allele of the homeobox gene Prox1 led to adult-onset obesity in mice resulting from abnormal lymph leakage from mispatterned and ruptured lymphatic vessels.8 • Parathyroid hormone–like hormone can inhibit adipogenesis and is downregulated in both active and chronic ophthalmopathy. This indicates the possibility of an increased risk of adipogenesis.18 • Adipogenesis in response to lymphatic fluid stasis is associated with a marked mononuclear cell inflammatory response.19 • Lymphatic fluid stasis potently upregulates the expression of fat differentiation markers both spatially and temporally.20 Clinicians often think that the swelling of a lymphedematous extremity results only from an accumulation of lymph fluid, which can be removed with noninvasive conservative regimens, such as complete decongestive therapy and controlled compression therapy.1 These therapies are efficacious if the excess swelling is in fact accumulated lymph, but they do not work if the excess volume is dominated by adipose tissue.21 The same may apply to microsurgical procedures employing lymphovenous shunts, transplantation of lymph vessels, and transfer of lymph nodes,22-26 which do not remove adipose tissue.

K23346_Neligan_32_Liposuction_r4_dc_0437-0446.indd 438

5/26/2015 10:01:41 AM

Chapter 32  Liposuction

439

Preoperative Assessment Volume Measurements The volumes of both extremities are always measured at each visit with water plethysmography, and the difference in volumes is designated as the excess volume.21,27,28

Venous Color Doppler Venous color Doppler examination is used to rule out any venous insufficiency, which can influence leg swelling.

Lymphoscintigraphy Lymphoscintigraphy provides good information not only on the anatomy, but also on the lymphatic function (transport). I use it mostly in patients with primary lymphedema and those with leg swelling of unknown origin, such as when lipedema is suspected.29

CT and MRI CT and MRI can detect enlarged lymph nodes and can show excess fat and edema. These modes can also be used to examine enlarged lymph nodes when primary or secondary malignancy is suspected.

Indications for Liposuction A surgical approach to remove the hypertrophied adipose tissue is appropriate when excess volume has not been achieved by conservative treatment and the patient has subjective discomfort from a heavy arm or leg.1 Liposuction should never be performed in a patient with a pitting edema, because it is dominated by accumulated lymph, which can be removed by conservative treatment.1 The primary and most significant goal is to transform a pitting edema into a nonpitting edema with conservative regimens such as complete decongestive therapy or controlled compression therapy. Pitting can be defined as a depression that is formed after pressure is exerted on the edematous tissue by the fingertip, which results in the squeezing of lymph into the surrounding tissue (Fig. 32-1, A). To standardize the pitting test, the surgeon presses as hard as possible with the thumb for 1 minute on the region under investigation. The amount of depression is estimated in millimeters. Swelling that is dominated by hypertrophied adipose tissue shows little or no pitting28 (Fig. 32-1, B). Approximately 4 to 5 mm of pitting in arm lymphedema and 6 to 8 mm of pitting in leg lymphedema are acceptable. Liposuction should not be done if pitting edema is present. This is because liposuction removes fat and not fluid, regardless if it theoretically could remove all of the accumulated fluid in a pitting lymphedema without excess adipose tissue formation.1 Liposuction also improves patients’ quality of life21,30 and reduces the incidence of erysipelas.31

K23346_Neligan_32_Liposuction_r4_dc_0437-0446.indd 439

5/26/2015 10:01:41 AM

440

Part V  Current Treatment of Lymphedema

A

FIG. 32-1  A, This woman has significant lymphedema of the arm after treatment for breast cancer. Pitting several centimeters in depth (arrows) (grade 1 edema) is seen. Arm swelling is dominated by the presence of fluid; that is, the accumulation of lymph. B, Another patient with pronounced arm lymphedema after treatment for breast cancer (grade 2 edema). No pitting is present despite hard pressure by the thumb for 1 minute. A slight reddening is visible at the two spots where pressure was exerted (arrows). The edema is completely dominated by the adipose tissue. The term edema is inaccurate at this stage, because the swelling is dominated by hypertrophied adipose tissue and not by lymph. At this stage, the aspirate contains either no or a minimal amount of lymph.

B

Surgical Technique Made-to-measure compression garments (two sleeves with a strap, two gauntlets, and two standard interim gloves) are measured and ordered 2 weeks before surgery.1 The healthy arm and hand are used as a template. One set (one sleeve and one standard interim glove) is sterilized and is put on the patient during surgery. I now use power-assisted liposuction. This facilitates liposuction, especially in the leg, which is more difficult to treat than the arm.1 The dry technique was initially used.32 Later, to minimize blood loss, a tourniquet was used combined with tumescence, which requires infiltration of 1 to 2 L of saline solution containing low-dose epinephrine and lignocaine.33,34 Through approximately 15 to 20 incisions, 3 mm long, liposuction is performed with 15 and 25 cm long cannulas with diameters of 3 and 4 mm (Fig. 32-2). After the arm distal to the tourniquet has been treated, a sterilized made-to-measure compression sleeve is applied (Jobst Elvarex–BSN Medical, compression class 2) to the arm to check bleeding and reduce postoperative edema. A sterilized Easy-Slide (Credenhill, England) helps to put on the garment and later is always used for this procedure. A sterilized, standard interim glove (Cicatrex interim, Thuasne, France), in which the tips of the fingers have been cut to aid gripping, is placed on the hand. The tourniquet is removed, and the most proximal part of the upper arm is treated with the tumescent technique.33,34

K23346_Neligan_32_Liposuction_r4_dc_0437-0446.indd 440

5/26/2015 10:01:42 AM

441

Chapter 32  Liposuction

C

D

B A

FIG. 32-2  Liposuction of arm lymphedema. The procedure takes about 2 hours. The preoperative to postoperative state is shown. Note the tourniquet, which has been removed in D, and the concomitant reactive hyperemia.

Finally, the proximal part of the compression sleeve is pulled up to compress the proximal part of the upper arm. The incisions are left open to drain through the sleeve. The arm is lightly wrapped with a large absorbent compress covering the entire arm (60 by 60 cm, Attends Cover Dri). The arm is maintained at the level of the heart on a large pillow. The compress is changed as needed.1 The next day, a made-to-measure gauntlet (a glove without fingers and thumb) (Jobst Elvarex– BSN Medical, compression class 2) is placed over the interim glove. The operating time averages 2 hours.1

Postoperative Care Garments are removed 2 days after surgery so the patient can shower. After showering the other set of garments is put on, and the used set is washed and dried. The patient repeats this step after another 2 days before discharge.1 The patient alternates between the two sets of garments (two sleeves, two gauntlets, and two interim gloves) during the 2 weeks after surgery. The patient changes garments daily or every other day so that a clean set is always worn after showering and lubricating the arm. After the 2-week control period, the garments are changed daily after they are washed. Washing “activates” the garments by increasing the compression caused by shrinkage.1

K23346_Neligan_32_Liposuction_r4_dc_0437-0446.indd 441

5/26/2015 10:01:42 AM

442

Part V  Current Treatment of Lymphedema

Controlled Compression Therapy To maintain the effect of liposuction and conservative treatment, a compression garment must be worn continuously.1,21,29 After compression therapy is started, the custom-made garment is altered at each visit with a sewing machine to compensate for the reduced elasticity and reduced arm volume. This procedure is most important during the first 3 months when the most significant changes in arm volume occur, but even later the garment must be adapted to compensate for wear and tear.1 Frequently the patient can manage this. At the 3-month visit, the arm is measured for new custom-made garments. This procedure is repeated at 6, 9, and 12 months.1 After complete reduction is achieved, sleeves without straps (for example, stay-up garments with silicone knobs) are ordered. If complete reduction has occurred by 6 months, the 9-month control may be omitted. If this happens, garments are prescribed for the next 6 months, which usually means double the amount that would be needed for 3 months. After the excess volume has decreased as much as possible, the treated arm typically becomes somewhat smaller than the normal arm, and a steady state is achieved. New garments can then be prescribed according to the latest measurements. In this way, the garments are renewed three or four times during the first year. Two sets of sleeve and glove garments are always available to the patient; one set is worn while the other set is washed. Thus a garment is worn continuously, and treatment is interrupted only briefly for showering and formal social occasions.1 The lifespan of the two garments worn alternately is usually 4 to 6 months. Complete reduction is usually accomplished after 3 to 6 months but frequently occurs earlier. After the first postoperative year, the patient is seen again after 6 months (1.5 years after surgery) and then at 2 years after surgery. The patient is then examined yearly only when new garments are prescribed for the upcoming year. Usually the patient receives four garments and four gloves (or four gauntlets). For active patients, six to eight garments and the same amount of gauntlets and gloves a year are needed. Patients without preoperative swelling of the hand can usually discontinue wearing the glove and gauntlet 6 to 12 months after surgery.1 For legs, I often use a maximum of two or three compression garments on top of each other, depending on the requirements to prevent pitting. A characteristic example is Elvarex compression class 3 (or 3 Forte) with a panty, Jobst Bellavar compression class 2 (or Elvarex compression class 2), and Elvarex compression class 2 (BSN Medical). The latter is a below-the-knee garment. Thus this patient needs two sets of two to three garments; one set is worn while the other set is washed. Depending on the age and activity of the patient, two such sets can last for 2 to 4 months. Thus these garments must be prescribed three to six times during the first year.1 After complete reduction has been achieved, no panty is needed, and stay-up garments (with silicone knobs) are ordered. The patient is examined yearly when all new garments are prescribed for the coming year.1

Treatment Outcomes and Complications Today chronic nonpitting arm lymphedema of up to 4 L in excess can be effectively removed by liposuction without any further reduction in lymph transport.29 Complete reduction is primar-

K23346_Neligan_32_Liposuction_r4_dc_0437-0446.indd 442

5/26/2015 10:01:42 AM

Chapter 32  Liposuction

443

ily achieved between 1 and 3 months. Long-term results have not shown any recurrence of the arm swelling21,29,35 (Fig. 32-3). Promising results can also be achieved for leg lymphedema (Fig. 32-4), for which complete reduction is usually reached at around 6 months.36-38 As with all liposuction procedures, numbness of the skin occurs postoperatively and usually disappears after a few months. No nerve damage or skin necrosis has occurred.

A

B

FIG. 32-3  A, This 74-year-old woman had had nonpitting arm lymphedema for 15 years after treatment for breast cancer. Preoperative excess volume was 3090 ml. B, Her postoperative result.

A

B

FIG. 32-4  A, This patient presented with primary lymphedema. The preoperative excess volume was 6630 ml. B, Her postoperative result, with complete reduction after 2 years.

K23346_Neligan_32_Liposuction_r4_dc_0437-0446.indd 443

5/26/2015 10:01:43 AM

444

Part V  Current Treatment of Lymphedema

Conclusion Accumulated lymph should be removed with well-documented conservative regimens until minimal or no pitting is seen. Any significant excess volume that remains can be removed with the use of liposuction, because neither further conservative treatment nor microsurgical reconstruction can remove the excess adipose tissue. A continuously worn compression garment will prevent recurrence. To date, I have trained and approved several teams. Recent publications from the Dutch and Scottish teams show the same favorable outcome as from our clinic.39,40

C linical P earls • Excess extremity swelling without pitting indicates that excess adipose tissue is present. • Adipose tissue can be removed with liposuction but not with conservative treatment or microsurgical procedures. • Lifelong use (24 hr/day) of custom-made, flat-knitted compression garments is—just as with conservative treatment—necessary to maintain the outcome of surgery. • Liposuction is a strong remedial in late-stage persistent primary or secondary lymphedema. A multidisciplinary team approach, including an occupational therapist and a physical therapist familiar with lymphedema treatment and measuring for compression garments, is important and should be centralized in a professional lymphedema center.

R EFERENCES 1. Brorson H. Surgical treatment of postmastectomy lymphedema—liposuction. In Lee BB, Bergan J, Rockson SG. Lymphedema: A Concise Compendium of Theory and Practice. Berlin: Springer, 2011. 2. Vague J, Fenasse R. Comparative anatomy of adipose tissue. In Renold AE, Cahill GF, eds. Handbook of Physiology, Section 5. Adipose Tissue. Washington, DC: American Physiology Society, 1965. 3. Ryan TJ. Lymphatics and adipose tissue. Clin Dermatol 13:493-498, 1995. 4. Mattacks CA, Sadler D, Pond CM. The control of lipolysis in perinodal and other adipocytes by lymph node and adipose tissue derived dendritic cells in rats. Adipocytes 1:43-56, 2005. 5. Pond CM. Adipose tissue and the immune system. Prostaglandins Leukot Essent Fatty Acids 73:17-30, 2005. 6. Borley NR, Mortensen NJ, Jewell DP, et al. The relationship between inflammatory and serosal connective tissue changes in ileal Crohn’s disease: evidence for a possible causative link. J Pathol 190:196-202, 2000. 7. Sadler D, Mattacks CA, Pond CM. Changes in adipocytes and dendritic cells in lymph node containing adipose depots during and after many weeks of mild inflammation. J Anat 207:769-781, 2005.

K23346_Neligan_32_Liposuction_r4_dc_0437-0446.indd 444

5/26/2015 10:01:43 AM

Chapter 32  Liposuction

445

8. Harvey NL, Srinivasan RS, Dillard ME, et al. Lymphatic vascular defects promoted by Prox1 haploinsufficiency cause adult-onset obesity. Nat Genet 37:1072-1081, 2005. 9. Schneider M, Conway EM, Carmeliet P. Lymph makes you fat. Nat Genet 37:1023-1024, 2005. 10. Jones B, Fishman EK, Hamilton SR, et al. Submucosal accumulation of fat in inflammatory bowel disease: CT/pathologic correlation. J Comput Assist Tomogr 10:759-763, 1986. 11. Sheehan AL, Warren BF, Gear MW, et al. Fat-wrapping in Crohn’s disease: pathological basis and relevance to surgical practice. Br J Surg 79:955-958, 1992. 12. Brorson H, Åberg M, Svensson H. Chronic lymphedema and adipocyte proliferation: clinical therapeutic implications. Lymphology 37(Suppl):153-155, 2004. 13. Lantz M, Vondrichova T, Parikh H, et al. Overexpression of immediate early genes in active Graves’ ophthalmopathy. J Clin Endocrinol Metab 90:4784-4791, 2005. 14. Bagheri S, Ohlin K, Olsson G, et al. Tissue tonometry before and after liposuction of arm lymphedema following breast cancer. Lymphat Res Biol 3:66-80, 2005. 15. Brorson H, Ohlin K, Olsson G, et al. Adipose tissue dominates chronic arm lymphedema following breast cancer: an analysis using volume rendered CT images. Lymphat Res Biol 4:199-210, 2006. 16. Brorson H. Adipose tissue in lymphedema: the ignorance of adipose tissue in lymphedema. Lymphology 37:135-137, 2004. 17. Brorson H, Ohlin K, Olsson G, et al. Breast cancer-related chronic arm lymphedema is associated with excess adipose and muscle tissue. Lymphat Res Biol 7:3-10, 2009. 18. Planck T, Parikh H, Brorson H, et al. Gene expression in Graves’ ophthalmopathy and arm lymphedema: similarities and differences. Thyroid 21:663-674, 2011. 19. Zampell JC, Aschen S, Weitman ES, et al. Regulation of adipogenesis by lymphatic fluid stasis: part I. Adipogenesis, fibrosis, and inflammation. Plast Reconstr Surg 129:825-834, 2012. 20. Aschen S, Zampell JC, Elhadad S, et al. Regulation of adipogenesis by lymphatic fluid stasis: part II. Expression of adipose differentiation genes. Plast Reconstr Surg 129:838-847, 2012. 21. Brorson H, Svensson H. Liposuction combined with controlled compression therapy reduces arm lymphedema more effectively than controlled compression therapy alone. Plast Reconstr Surg 102:10581067; discussion 1068, 1998. 22. Baumeister RG, Siuda S. Treatment of lymphedemas by microsurgical lymphatic grafting: what is proved? Plast Reconstr Surg 85:64-74; discussion 75-76, 1990. 23. Baumeister RG, Frick A. The microsurgical lymph vessel transplantation. Handchir Mikrochir Plast Chir 35:202-209, 2003. 24. Campisi C, Davini D, Bellini C, et al. Lymphatic microsurgery for the treatment of lymphedema. Microsurgery 26:65-69, 2006. 25. Saaristo AM, Niemi TS, Viitanen TP, et al. Microvascular breast reconstruction and lymph node transfer for postmastectomy lymphedema patients. Ann Surg 255:468-473, 2012. 26. Viitanen TP, Visuri MP, Hartiala P, et al. Lymphatic vessel function and lymphatic growth factor secretion after microvascular lymph node transfer in lymphedema patients. Plast Reconstr Surg Glob Open 1:1-9, 2013. 27. Brorson H, Svensson H. Complete reduction of lymphoedema of the arm by liposuction after breast cancer. Scand J Plast Reconstr Surg Hand Surg 31:137-143, 1997. 28. Brorson H. Liposuction in arm lymphedema treatment. Scand J Surg 92:287-295, 2003. 29. Brorson H, Svensson H, Norrgren K, et al. Liposuction reduces arm lymphedema without significantly altering the already impaired lymph transport. Lymphology 31:156-172, 1998. 30. Brorson H, Ohlin K, Olsson G, et al. Quality of life following liposuction and conservative treatment of arm lymphedema. Lymphology 39:8-25, 2006. 31. Brorson H, Svensson H. Skin blood flow of the lymphedematous arm before and after liposuction. Lymphology 30:165-172, 1997.

K23346_Neligan_32_Liposuction_r4_dc_0437-0446.indd 445

5/26/2015 10:01:43 AM

446

Part V  Current Treatment of Lymphedema

3 2. Clayton DN, Clayton JN, Lindley TS, et al. Large volume lipoplasty. Clin Plast Surg 16:305-312, 1989. 33. Klein JA. The tumescent technique for liposuction surgery. Am J Cosm Surg 4:263-267, 1987. 34. Wojnikow S, Malm J, Brorson H. Use of a tourniquet with and without adrenaline reduces blood loss during liposuction for lymphoedema of the arm. Scand J Plast Reconstr Surg Hand Surg 41:243-249, 2007. 35. Brorson H, Freccero C, Ohlin K, et al. Seventeen years’ experience of complete reduction of arm lymphedema following breast cancer. Lymphology 45(Suppl):279-281, 2012. 36. Brorson H, Ohlin K, Svensson B, et al. Controlled compression therapy and liposuction treatment for lower extremity lymphedema. Lymphology 41:52-63, 2008. 37. Brorson H, Freccero C, Ohlin K, et al. Liposuction normalizes elephantiasis of the leg—a prospective study with an eight-year follow-up. Lymphology 45(Suppl):292-295, 2012. 38. Brorson H, Ohlin K, Svensson B, et al. Controlled compression therapy and liposuction treatment for lower extremity lymphedema. Lymphology 41:52-63, 2008. 39. Damstra RJ, Voesten HG, Klinkert P, et al. Circumferential suction-assisted lipectomy for lymphedema after surgery for breast cancer. Br J Surg 96:859-864, 2009. 40. Schaverien MV, Munro KJ, Baker PA, et al. Liposuction for chronic lymphoedema of the upper limb: 5 years of experience. J Plast Reconstr Aesthet Surg 65:935-942, 2012.

K23346_Neligan_32_Liposuction_r4_dc_0437-0446.indd 446

5/26/2015 10:01:43 AM

C hapter 33 Multiple Lymphaticovenous Anastomoses and Multiple Lymphatic-Venous-Lymphatic Anastomoses

ML

Corrado Cesare Campisi, Melissa Ryan, Corradino Campisi

K ey P oints • Microsurgical derivative and reconstructive lymphaticovenous anastomoses (multiple lymphaticovenous anastomoses and multiple lymphatic-venous-lymphatic anastomoses) can provide an effective functional repair of the lymphatic pathway by diverting the lymph into adjacent veins proximal to the source of the lymphatic obstruction. • Lymphatic microsurgery is appropriate for both primary and secondary obstructive lymphedemas and as a preventive approach during other general surgeries in which there is the risk of disrupting the lymphatic pathways. • Lymphatic microsurgery is most effective when it is performed early in the disease course and in conjunction with a combined treatment protocol such as complete lymphedema functional therapy (CLyFT). • Advanced stages of lymphedema can be appropriately treated with multiple lymphaticovenous anastomoses or multiple lymphatic-venous-lymphatic anastomoses to relieve lymph stasis but may require a complementary procedure to remove residual excess fibrous and adipose tissue contributing to poor lymphatic drainage.

Chronic lymphedema is a progressive and relatively painless swelling of any peripheral tissue, including the limbs, head and neck, breast, trunk, or genitals; it results from a reduced transport capacity of the lymphatic system. Chronic lymphedema can be classified as primary or secondary, depending on the cause. In patients with secondary lymphedema, a specific external cause (filariasis, previous surgery, radiation, malignancy, infection or inflammation, or trauma) can be identified and is believed to impact on a presumed previously normal functioning lymphatic system by causing an obstruction to the lymphatic flow. Most of the clinical conditions that are considered primary lymphedema are the result of truncal lymphatic malformations that develop during the final stages of lymphovasculogenesis1,2 when the lymphatic trunks, vessels, and nodes are formed.3,4 These malformations result in hypoplasia, 447

K23349_Neligan_33_Multi Lymph_r4_dc_0447-0462.indd 447

5/27/2015 9:38:14 AM

448

Part V  Current Treatment of Lymphedema

BOX 33-1  Staging of Lymphedema Based on Immunohistologic Criteria, Lymphoscintigraphic Findings, Clinical Symptoms, and Degree of Physical Disability Stage 1 A Latent lymphedema without clinical evidence of edema but with impaired lymph transport capacity (provable by lymphoscintigraphy) and initial immunohistochemical alterations of lymph nodes, lymph vessels, and extracellular matrix B Initial lymphedema, totally or partially decreasing by rest and draining position, with worsening impairment of lymph transport capacity and immunohistochemical alterations of lymph collectors, nodes, and extracellular matrix

Stage 2 A Increasing lymphedema with vanishing lymph transport capacity, relapsing lymphangitic attacks, fibroindurative skin changes, and developing disability B Column-shaped limb fibrolymphedema with lymphostatic skin changes, suppressed lymph transport capacity, and worsening disability

Stage 3 A Properly called elephantiasis, with scleroindurative pachydermatis, papillomatous lymphostatic verrucosis, and no lymph transport B Extreme elephantiasis with total disability

hyperplasia, or aplasia of the lymphatic vessels and/or lymph nodes and may clinically manifest as an obstruction or dilation. To provide a comprehensive classification system of lymphedema that encompasses the immunohistopathologic criteria, level of clinically evident edema, lymphoscintigraphic findings, and level of physical disability, we developed a three-stage model5,6 (Box 33-1). In clinical practice, stages 1A, 1B, 2A, and 2B can be considered early manifestations of disease and stages 3A and 3B (usually known as elephantiasis) are chronic and advanced stages. Lymphedema is a progressive disease and can move rapidly between the stages without adequate treatment.7-10

Preoperative Considerations Diagnosis The diagnosis of lymphedema and other lymphatic disorders should be performed by clinicians and lymphologists experienced in this area. The essential diagnostic criterion is evidence of slow, reduced, or completely absent lymphatic flow in the tissue affected by the swelling. Lymphoscintigraphy is generally considered the benchmark procedure to measure this flow, although procedures have not been standardized. In Genoa, Italy, lymphoscintigraphy, which is performed with either technetium-99m (99mTc)-labeled antimony sulfur colloid or 99mTc-nanocolloid human serum albumin (90% of the particles are greater than 80 nm), is used in the diagnostic workup before surgery to determine the eligibility for derivative multiple lymphaticovenous anastomoses (MLVA). Lymphoscintigraphy clearly indicates whether the edema has a lymphatic origin in patients and provides useful data about the etiologic factors and pathohistologic nature of the lymphedema. A transport index is calculated to categorize the lymphatic flow as normal or pathologic (Fig. 33-1).

K23349_Neligan_33_Multi Lymph_r4_dc_0447-0462.indd 448

5/27/2015 9:38:14 AM

Chapter 33  MLVA and MLVLA

449

TRANSPORT INDEX TI 5 K 1 D 1 (0.04 3 T) 1 N 1 V K 5 Transport kinetics 0 Normal 3 Mild delay 5 Marked delay 9 No transport D 5 Distribution of the tracer 0 Normal 3 Mild dermal diffusion 5 Marked dermal diffusion 9 Absent visualization T 5 Appearance time of lymph nodes (min) N 5 Visualization of lymph nodes 0 Normal 3 Mild 5 Poor 9 Absent V 5 Visualization of lymph vessels 0 Normal 3 Mild 5 Poor 9 Absent

FIG. 33-1  Lymphoscintigraphy transport index calculation. The normal transport index is less than 10, and a pathologic transport index is greater than 10. (TI, Transport index.)

A score of less than 10 signifies a normal transport index, and a score equal to or greater than 10 signifies a pathologic transport index. Scores are determined bilaterally, even in patients with unilateral swelling. (Other diagnostic and preoperative considerations are discussed in the Surgical Techniques section.)

Surgical Techniques The first use of microsurgery for the treatment of lymphatic disorders was in 1962 when Cockett and Goodwin11 performed an anastomosis of a dilated lumbar lymphatic vessel to the spermatic vein to treat a patient with chyluria. As a result of this first procedure, subsequent developments in microsurgical techniques have enabled lymphaticovenous anastomosis to emerge as a feasible and relatively commonplace treatment option for lymphedema. In 1977 and 1979 O’Brien et al12,13 described their use of microlymphatic surgery for the treatment of secondary obstructive lymphedema. They created three lymphaticovenous anastomoses at or above the elbow (or knee) and observed a reduced incidence of cellulitis after surgery. Their microlymphatic techniques were applicable to both upper and lower limbs and were also used for localized cases of obstructive lymphedema after trauma and congenital constriction bands. They emphasized that considerable experience in microvascular surgery was required to perform the procedure. The field was further advanced in 1981 when Degni14,15 introduced an original technique of lymphaticovenous anastomosis to divert the lymph into a vein in patients with blocked lymphatic vessels, particularly when lymphographic findings showed good function and intact permeability of lymphatic vessels. At almost the same time, Clodius et al16,17 stated that microsurgery for

K23349_Neligan_33_Multi Lymph_r4_dc_0447-0462.indd 449

5/27/2015 9:38:14 AM

450

Part V  Current Treatment of Lymphedema

primary and secondary lymphedema, which consisted of shunts between the lymphatic vessels and veins, was a well-established surgical technique. However, problems arose from irreversible changes in the peripheral lymphatic system and connective tissues and from the obliteration of the deep lymphatics best suited for lymphaticovenous anastomoses in more chronic lymphedema. Therefore Clodius cautioned that lymphaticovenous shunts should be performed early before the appearance of fibrotic tissue changes. Despite advances in microsurgery, the most suitable operation for peripheral lymphedema remains unclear.

Derivative Approaches The first derivative microsurgical procedures used lymph nodal–venous shunts. These have largely been abandoned because of the high rate of anastomotic closures.18 Lymph nodal pulp has a thrombogenic effect on the venous blood, and there is also a frequent reendothelialization of the lymph nodal surface, both of which contribute to anastomotic closure. Worldwide, surgeons have moved to the anastomosis of lymphatic vessels directly into the veins to increase the long-term efficacy of the procedures.19,20 More recently, our preferred technique has been telescopic MLVA. Lymphaticovenous anastomoses involve joining an appropriate lymphatic vessel to a collateral branch of a main vein and ensuring that the vein has competent valvular function and continence. This is essential to prevent reflux of the blood into the lymphatic vessel and thereby thrombosis of the anastomosis.21 Anastomoses that are created in close proximity to a valve in the vein are able to overcome the difference in pressure between the venous and lymphatic systems. In this way the valvular pumping creates a suction that pulls the lymph immediately through the anastomosis to prevent thrombosis. The Center of Lymphatic Surgery and Microsurgery at the University of Genoa, Italy, has obtained excellent stable clinical outcomes for 40 years with the use of the MLVA and multiple lymphatic-venous-lymphatic anastomoses (MLVLA)5,22,23 (Fig. 33-2). Anastomoses are performed at a single site; larger lymphatic vessels are attached to collateral branches of the

FIG. 33-2  The derivative MLVA and reconstructive MLVLA technique with the interposition of an autologous vein graft between the lymphatics above and below the obstacle to the lymph flow.

K23349_Neligan_33_Multi Lymph_r4_dc_0447-0462.indd 450

5/27/2015 9:38:14 AM

Chapter 33  MLVA and MLVLA

451

FIG. 33-3  The passage of blue lymph into the vein branch can be seen under the operating microscope, which verifies the patency of the anastomosis.

main veins close to vein valves to avoid backflow of blood and the closure of the anastomosis. A single-site approach also minimizes the number of incisions and thereby potential entry sites for infection. Blue patent violet (BPV) dye, a sodium or calcium salt of diethyl ammonium hydroxide, is used intraoperatively to identify properly functioning lymphatic vessels at the surgical incision. Healthy appearing lymphatics found at the surgical site are directly introduced into the vein by a U-shaped stitch and then fixed to the vein cut end by means of additional stitches between the vein border and perilymphatic adipose tissue. We create the MLVA by using lymphatic vessels with the perilymphatic tissue intact. This ensures that these vessels are functioning and also helps to prevent thrombosis. At the end of the procedure, the first U-shaped stitch is removed to avoid the risk of closure of the lymphatic collectors. With the use of BPV dye, properly functioning lymphatics appear blue, and the passage of blue lymph into the vein branch verifies the patency of the lymphaticovenous anastomosis under the operating microscope when the anastomosis is completed (Fig. 33-3). For lower limb lymphedema, the MLVAs are created at the site of a single incision made at the subinguinal region. For upper limb lymphedema, MLVAs are created in the middle third of the volar surface of the arm. Both surface and deep afferent lymphatic collectors that are visualized by the BPV dye are used. The deep lymphatics are generally located between the humeral artery, vein, and median nerve and introduced by a telescopic technique into a patent branch of one of the humeral veins containing a functioning valve.

Multiple Lymphaticovenous Anastomoses It is possible to perform multiple lymphaticovenous anastomoses at the same time; the most important requirement is that the vein branch be patent. Duplex scanning is performed in all patients to identify any venous disorders that may contribute to the edema. In most patients, it is possible to correct these venous dysfunctions at the same time as the microsurgery. This is done by performing procedures, such as valvuloplasty, for venous insufficiency with 6-0 nylon sutures. In a few cases, venous disease is an indication for performing a venous graft between the lymphatic collectors above and below the site of lymphatic obstruction, because the valvular incompetence of the diseased veins compromises the efficacy of the anastomoses if MLVAs were

K23349_Neligan_33_Multi Lymph_r4_dc_0447-0462.indd 451

5/27/2015 9:38:14 AM

452

Part V  Current Treatment of Lymphedema

performed instead. This venous bridge type of graft is called a lymphatic-venous-lymphatic anastomosis. Competent venous segments are harvested from the same operative site or can be taken from the forearm (usually the cephalic vein). The length of the grafted vein section varies from 7 to 15 cm, as needed. It is essential to gather several lymphatic collectors to join to the distal end of the vein to ensure that the vein segment is filled with sufficient lymph to avoid closure from subsequent fibrosis. The competent valves of the vein segment are vital to direct the flow of lymph in the correct direction and to avoid gravitational backflow or reflux. As with the lymphaticovenous anastomosis, the lymphatic collectors are directly introduced into the vein cut ends by means of a U-shaped stitch, which is then stabilized with additional peripheral stitches and ultimately removed as described previously.

Clinical Experience Between 1973 and February 2014, 3007 patients with upper and lower limb lymphedema and primary and secondary disease received microsurgical treatment for peripheral lymphedema in Genoa, Italy. Patients were followed for a minimum of 5 years, and our maximum follow-up was 25 years after surgery. Our routine follow-up consisted of periodic clinical evaluations at 1, 3, 6, and 12 months after microsurgery and then annually for a minimum of 5 years. This close patient follow-up provided optimal control for the long-term clinical outcome. Histologic samples taken during surgery showed that primary lymphedemas typically involved lymph nodal dysplasias (leukocyte adhesion deficiency II according to Papendieck’s classifications24) with hypoplastic lymph nodes associated with sinus histiocytosis and a thick fibrous capsule and microlymphangioadenomyomatosis. These dysplasias caused an obstruction to lymphatic flow that was evident in the changes in the afferent lymphatic collectors. These were dilated, swollen, and tortuous, with thickened walls and reduced numbers of smooth muscle cells, which were fragmented by numerous fibrotic elements. In our clinical experience, secondary lymphedemas are usually related to lymphadenectomy and radiotherapy as part of oncologic treatment (carcinoma of the breast, uterus, penis, bladder, prostate gland, rectum, and seminoma of the epididymis) or to complications from minor surgeries for varicose veins, crural and inguinal hernias, lipomas, tendinous cysts, or inguinal and axillary lymph nodal biopsies. Most of the lymphedemas treated were at stages 2A (38%) and 2B (53%), with 3.5% at stage 1B and 5.5% at stages 3A and 3B, according to the Campisi staging system for lymphedema25 (see Box 33-1). Objective clinical measures of lymphedema consisted of limb water volumetry, circumferences, and lymphoscintigraphy. Lymphoscintigraphy, which was performed with either 99mTc-labeled antimony sulfur colloid or 99mTc-nanocolloid human serum albumin (90% of the particles were greater than 80 nm), was used in the diagnostic workup before surgery to determine the eligibility for derivative MLVA. Lymphoscintigraphy clearly indicated whether the edema had a lymphatic origin in patients and provided useful data about the cause and pathohistologic nature of the

K23349_Neligan_33_Multi Lymph_r4_dc_0447-0462.indd 452

5/27/2015 9:38:14 AM

Chapter 33  MLVA and MLVLA

453

lymphedema. Lymphoscintigraphy was also used to verify the patency of the microanastomoses in the long term by direct and indirect methods (Fig. 33-4). These included the following: • Reduced dermal backflow of the tracer and the appearance of preferential lymphatic pathways not evident preoperatively • The disappearance of the tracer at the site of the lymphaticovenous anastomoses, indicating the passage of lymph into the bloodstream • Earlier liver uptake of tracer compared with preoperative parameters, indicating indirect evidence of the passage of lymph in the bloodstream Most recently, the evaluation of the superficial lymphatic pathways distal to the surgical site in the affected limb has also been performed with the photodynamic eye (PDE) method of injection of indocyanine green fluorescence. Simultaneous use of PDE with BPV provides two methods of visualizing the lymphatic flow and confirming the patency of the anastomoses intraoperatively.26,27 PDE can be used after surgery, in addition to lymphoscintigrapy, to verify the long-term stability of the surgical outcomes.

A

B

C

D

FIG. 33-4  Preoperative (A and C) and postoperative (B and D) lymphoscintigraphy of the upper limbs in a patient treated with MLVA. Note the appearance of preferential lymphatic pathways after surgery. The arrows show the lymphatic flow in the right arm. In B and D, the areas of greater lymphatic flow are indicated by a thicker white line and a concentration of white pixels corresponding to the lymph node(s), providing evidence of the new lymphatic pathways that were created surgically in that arm.

K23349_Neligan_33_Multi Lymph_r4_dc_0447-0462.indd 453

5/27/2015 9:38:15 AM

454

Part V  Current Treatment of Lymphedema

In our series, compared with preoperative conditions, all patients obtained significant reductions in excess limb volume of more than 85%, with an average of 70% as measured by limb water volumetry and circumference. These results were stable over an average of 10 years of follow-up. Outcomes improved with earlier microsurgical intervention because of no or minimal fibrosclerotic tissue changes in the lymph vessel walls and surrounding tissues. More than 85% of patients with earlier stages of disease (stages 1 or 2A and 2B) progressively stopped using conservative therapies over the follow-up period (Fig. 33-5). Forty-three percent of patients with more advanced lymphedema (stages 3A and 3B) decreased the frequency of physical therapy in the long term (Fig. 33-6). Infective rates, particularly lymphangitis, cellulitis, and erysipelas, dropped by 91% compared with preoperative conditions.

A

B

FIG. 33-5  A, This woman is seen preoperatively with lymphedema of the left arm (stages 2A and 2B) after mastectomy. B, Follow-up 5 years after surgery.

A

B

FIG. 33-6  A, This man had elephantiasis before surgery. B, Long-term stable results are shown after MLVA and appropriate weight control at 5-year follow-up.

K23349_Neligan_33_Multi Lymph_r4_dc_0447-0462.indd 454

5/27/2015 9:38:15 AM

455

Chapter 33  MLVA and MLVLA

Additional Considerations Patients Refractory lymphedema unresponsive to conservative treatment measures may be appropriately managed by surgical means (Fig. 33-7). Indications for surgery included insufficient volume reduction by appropriate conservative methods (less than 50% reduction), recurrent episodes of lymphangitis or erysipelas, intractable pain or discomfort usually associated with the excess swelling and inflammation, loss of limb function and increasing disability, and patient dissatisfaction with previous treatment outcomes and willingness to proceed with surgery. Relative contraindications to lymphatic microsurgery were few but included lymphatic-lymph node aplasia (exceedingly rare), diffuse metastatic carcinomas, and extremely advanced lymphedema (stage 3B) unresponsive to conservative measures. The ideal indications for surgery were represented by the following: (1) early stages (1B, 2A, and 2B) of disease, (2) lymphoscintigraphy showing a low inguinal or axillary lymph nodal uptake of radioactive tracer and minimal or no passage of the tracer beyond this proximal nodal area, (3) excellent patient compliance, and (4) a lymphologic center where the patient was easily referred for any needs, in addition to the Center of Lymphatic Surgery, where the patient underwent this specialized surgery.

Combined Treatment Protocol Patients were treated in Genoa under a prescribed treatment protocol: complete lymphedema functional treatment (CLyFT)26 (Fig. 33-8). This protocol consisted of intensive physical therapy

100 Normal Stage 1A-1B Stage 2A Stage 2B Stage 3A-3B

75 50 25 0

Diagnosis

6 mo-1 yr 3-5 yr 10-15 yr

FIG. 33-7  Long-term reduction in edema volume after lymphatic microsurgery according to the stage of disease (p .0.05).

Phase 1 • CPT •  6-12 mo

Phase 2 • Microsurgery •  1 wk

Phase 3 •  Postoperative rehabilitation •  5 yr follow-up

FIG. 33-8  CLyFT protocol for lymphedema. (CPT, Comprehensive physical therapy.)

K23349_Neligan_33_Multi Lymph_r4_dc_0447-0462.indd 455

5/27/2015 9:38:15 AM

456

Part V  Current Treatment of Lymphedema

with uniform and sequential intermittent pressure devices and manual and mechanical lymphatic drainage. Intermittent negative pressure therapy was also included, because it improved microperfusion and venous and lymphatic flow in the lower limbs.28 When it was added to a combined treatment protocol for lymphedema, intermittent negative pressure therapy provided additional benefits in volume reduction. All patients wore compression garments or had multilayered functional bandaging to maintain the volume reductions obtained by the CLyFT protocol and to prepare the limb for surgery. CLyFT was applied in three phases: (1) an intensive preoperative phase to reduce the size of the affected limb as much as possible before the microsurgical intervention; (2) a gentle postoperative phase in which the pressure of the lymphatic drainage was gradually increased as healing continued; and (3) a long-term maintenance phase of daily mechanical lymphatic drainage and physical exercise (particularly swimming) to strengthen the anastomoses over time. After surgery, all patients were prescribed antithrombotic medications (low-molecular-weight heparin immediately after surgery followed by low doses of aspirin for 1 to 2 years) to prevent thrombosis. Starting the mechanical drainage elements of CLyFT within 1 week of surgery helped to develop the flow of lymph through the anastomoses29 and was effective in maintaining the long-term patency of the anastomoses. Patients received antibiotic therapy (amoxicillin for the first week and then long-acting penicillin).

Timing of Surgery The timing of surgery was based on the clinical indications; in the initial stages of lymphedema, microsurgery was performed early in the treatment process before the progression to fibrosclerotic tissue changes and excess adipose deposition occurred. These patients (stages 1B, 2A, and 2B) had 1 or 2 weeks of CLyFT to minimize the edema before surgery. In more advanced stages of lymphedema (stages 3A and 3B, usually known as elephantiasis), in which these fibrotic tissue changes had already occurred, microsurgery was performed when the physical therapy component of CLyFT failed to obtain further edema reduction or prevent recurrent lymphangitis. This was some weeks or months later. Lymphatic microsurgery done at this time allowed further amelioration in volume reduction and pathology.30-32 We follow a prescribed algorithm for the treatment of lymphatic disorders (Fig. 33-9). Emphasis is placed on the prevention of lymphatic injuries through safe and effective surgical techniques. The treatment of lymphatic disorders, with particular emphasis on lymphedema, is based on the stage of disease. Early stages are treated as soon as possible with MLVA or MLVLA as appropriate.33-39 Advanced stages of disease are treated with MLVA or MLVLA but often require additional surgical treatment to remove the fibrotic tissues that occur and further impede lymphatic flow. The rationale behind the prevention of lymphatic injuries and the treatment of advanced lymphedema will be discussed next.

Prevention of Lymphatic Injuries The lymphatic microsurgical preventive healing approach (LYMPHA) is discussed in detail in Chapter 40 (Fig. 33-10).

K23349_Neligan_33_Multi Lymph_r4_dc_0447-0462.indd 456

5/27/2015 9:38:15 AM

457

Chapter 33  MLVA and MLVLA

INDICATIONS FOR LYMPHATIC SURGERY ACCORDING TO CLyFT PROTOCOL Lymphatic disorders

Prevention

To avoid lymphatic injuries

Treatment

LYMPHA

Early

MLVA

Late

MLVLA

FLLA-LVSP

FIG. 33-9  Algorithm for the treatment of lymphatic disorders. (LYMPHA, Lymphatic microsurgical preventive healing approach; MLVA, multiple lymphaticovenous anastomoses; MLVLA, multiple lymphatic-venous-lymphatic anastomoses; FLLA-LVSP, fibro-lipo-lympho-aspiration by lymph vessel–sparing procedures.)

FIG. 33-10  LYMPHA technique. Blue dyed lymphatics from the upper limb are anastomosed to a collector branch of the axillary or thoracodorsal vein.

Advanced Lymphedema Microsurgery has an important role in the treatment of advanced and chronic lymphedema, because it helps to resolve the lymph stasis that contributes markedly to the chronic swelling associated with advanced lymphedema. As with earlier stages of lymphedema, advanced lymphedema is the result of obstacles to the flow of lymph, generally at the level of the lymph nodes, although over time all lymphatic vessels become dilated and less efficient, allowing lymph to leak into the tissues. Chronic lymphedema is characterized by a buildup of hard, fibrotic tissue that contributes to the slow or almost static lymphatic flow. Recent mouse models show the physiologic effects of this lymph stasis in both primary (lymphatic malformations) and secondary (obstructive) lymphedemas.10 Lymph stasis in models of secondary lymphedema (for example, from damage to lymph nodes in mouse tails) leads to hyperplasia of lymph channels in the tail as a response to increased fluid. These hyperplastic channels are less efficient and leak fluid, further increasing the swelling.40 Chronic lymph stasis is also related to a poor immune response; lymph stasis is also implicated in decreased immune cell trafficking,41 whereas dendritic cells accumulate in the der-

K23349_Neligan_33_Multi Lymph_r4_dc_0447-0462.indd 457

5/27/2015 9:38:16 AM

458

Part V  Current Treatment of Lymphedema

mis and fail to migrate as usual to the nodes, where they play a role in antigen presentation.42 It is well known that a high risk of frequent infections occurs with advanced stages of lymphedema, and this appears to be related to chronic lymph stasis. Therefore the primary treatment objective, regardless of stage, is to reduce this lymph stasis through an improvement in the lymph transport capacity.43 For the patient with chronic fibrosclerosis, the initial objective is the same as that for patients in the early stages: to improve the lymph transport capacity with reconstructive microsurgery and to reduce swelling. However, what is more important is to help mediate the immune response within the lymphedematous limb. If the lymph stasis remains, there is an increased risk of infection and inflammation, which further damage the fluid transport. For this reason, microsurgical techniques (MLVA/MLVLA) are used to improve the lymph stasis that is also in advanced stages of lymphedema. Thus a secondary goal is to remove the fibrous tissue. Treatments that focus solely on reducing the swelling in chronic lymphedema through tissue remodeling, such as conservative treatment or liposuction, do not address the key factor in lymphedema—lymph stasis—and therefore are likely to have only shortterm benefits.

Fibro-lipo-lympho-aspiration With the Lymph Vessel–Sparing Procedure Even in patients with advanced lymphedema who have prevalent lymph stasis but little fibrotic tissue, it may be sufficient to perform a functional repair of the lymphatic route with MLVA to resolve the lymph stasis and therefore the lymphedema. However, in patients with significant fibrosis and excess adipose tissue, this excess tissue serves as a continual residual impediment to lymphatic flow and an ongoing risk of infection. There is some residual blockage in flow with excess fibrotic and adipose tissue resulting from the tissue blocking the superficial lymphatic pathways, even in those patients after MLVA treatment in whom lymphoscintigraphy and the PDE test confirm the patency of the anastomoses. The removal of the fibrotic tissue is a secondary goal of treatment.22 Recently liposuction has been advanced as a treatment to remove fibrotic and excess adipose tissue for advanced stages of lymphedema. Reductions in the size of the limb are much more aesthetically pleasing than those achieved with an excisional technique such as the Charles procedure.44 Compared with the Charles procedure, liposuction is less invasive, but caution is still required when the procedure is used in patients with lymphedema.45,46 Because of their already damaged lymphatic system, care must be taken during liposuction to avoid further injury to the lymph nodes and vessels. Investigations of lymph vessels in cadavers after dry and tumescent liposuction showed that a tumescent procedure and a parallel approach were necessary to avoid injury, and this was in people with a normal lymphatic system.45 In patients with lymphedema, the lymph vessels and channels are often dilated and tortuous in the advanced stages and therefore may be more difficult to avoid with the liposuction cannula.47 For this reason, the fibro-lipo-lymphoaspiration with the lymph vessel–sparing procedure (FLLA-LVSP) was developed to spare the lymphatic pathways already reestablished by lymphatic microsurgery22 (Figs. 33-11 and 33-12). The FLLA-LVSP differs from standard liposuction for lymphedema in two ways. Intraoperatively, the lymphatic pathways are visualized by BPV and fluorescent dyes (indocyanine green) so that these can be avoided with the liposuction cannula. All excess tissue not immediately adjacent to the lymphatic channels can be aspirated without risk to lymphatic structures. This differs from

K23349_Neligan_33_Multi Lymph_r4_dc_0447-0462.indd 458

5/27/2015 9:38:16 AM

459

Chapter 33  MLVA and MLVLA

A

B

FIG. 33-11  A, Bilateral lower limb lymphedema. B, Postoperative result of FLLA-LVSP in a patient already treated with MLVA.

A

B

FIG. 33-12  A, Right lower leg lymphedema (stage 3A) before surgery. B, Immediate postoperative result of FLLA-LVSP in a patient already treated by MLVA.

ordinary liposuction where as much tissue as possible is removed.48,49 The goal of FLLA-LVSP is to complement the MLVA by increasing the lymphatic flow. We postulate that removal of the excess tissue eliminates all remaining tissue impediment to lymph flow, thus improving lymph stasis. In time, there is the possibility of stabilizing the balance of lymph in and out of the limb. As a result, the use of compressive garments and conservative treatments is reduced or even completely withdrawn. Other liposuction techniques, which do not treat the lymph stasis, require the continual use of compression garments to prevent reswelling of the limb.22,48,49

K23349_Neligan_33_Multi Lymph_r4_dc_0447-0462.indd 459

5/27/2015 9:38:16 AM

460

Part V  Current Treatment of Lymphedema

Conclusion Large robust samples and long-term follow-up periods have established lymphatic microsurgery as an effective method for treating lymphatic obstruction in lymphedema. MLVA and MLVLA techniques, when performed at a single site, are postulated as the treatment of choice for patients in the initial stages of disease in which a functional repair to the lymphatic flow can be achieved that is stable and consistent for up to 25 years after surgery. MLVA can be used preventively in most surgeries in which there is a risk of damage to lymphatic structures to allow continuity in lymphatic flow. MLVA is also the mainstay of treatment in our practice for advanced lymphedema to reestablish the lymphatic flow in the affected limb and can be complemented with the FLLA-LVST to remove the excess tissue creating a residual barrier to this flow.

C linical P earls • The use of lymphoscintigraphy is invaluable in diagnosis and can also be used to detect disturbances in lymphatic flow in an affected limb before clinical evidence of swelling (stage 1A). In such cases, preventive microsurgery can be performed to prevent the progression of lymph stasis and evident lymphedema (LYMPHA). • For comorbid vein disease that requires the use of lymphatic-venous-lymphatic anastomoses to bridge the lymphatic obstruction with an autologous vein graft, we anastomose the proximal stump of the vein graft with lymphatic vessels isolated above the obstruction and distal stump with lymph collectors isolated below. From the physiodynamic viewpoint, the most important aspect to achieve an effectively draining vein interposition graft is to anastomose a greater number of lymph collectors distally than proximally. In this way, a positive pressure and antigravitational lymphatic-venous-lymphatic pressure gradient is created. • Lymphatic microsurgery is applicable to the treatment of lymphedema regardless of cause or stage (except for stage 3B, which has no detectable flow of lymph and is resistant to conservative treatments). By now, decades of clinical studies have demonstrated its effectiveness. However, the best results are obtained in the earlier stages of disease before adverse tissue changes occur. For this reason, we urge clinicians not to reserve microsurgery as a secondary treatment but to consider its application as soon as possible within the treatment course and even as a prevention method.

R EFERENCES 1. Rutkowski JM, Boardman KC, Swartz MA. Characterization of lymphangiogenesis in a model of adult skin regeneration. Am J Physiol Heart Circ Physiol 291:H1402-H1410, 2006. 2. Goldman J, Le TX, Skobe M, et al. Overexpression of VEGF-C causes transient lymphatic hyperplasia but not increased lymphangiogenesis in regenerating skin. Circ Res 96:1193-1199; comment, 1132-1134, 2005. 3. Bastide G, Lefebvre D. Anatomy and organogenesis and vascular malformations. In Belov S, Loose DA, Weber J, eds. Vascular Malformations. Reinbek, Germany: Einhorn-Presse Verlag, 1989. 4. Leu HJ. Pathoanatomy of congenital vascular malformations. In Belov S, Loose DA, Weber J, eds. Vascular Malformations. Reinbek, Germany: Einhorn-Presse Verlag, 1989.

K23349_Neligan_33_Multi Lymph_r4_dc_0447-0462.indd 460

5/27/2015 9:38:16 AM

Chapter 33  MLVA and MLVLA

461

5. Murdaca G, Cagnati P, Gulli R, et al. Current views on diagnostic approach and treatment of lymphedema. Am J Med 125:134-140, 2012. 6. Campisi C, Boccardo F. Microsurgical techniques for lymphedema treatment: derivative lymphaticvenous microsurgery. World J Surg 28:609-613, 2004. 7. Schneider M, Conway EM, Carmeliet P. Lymph makes you fat. Nat Genet 10:1023-1024, 2005. 8. Rutkowski JM, Davis KE, Scherer PE. Mechanisms of obesity and related pathologies: the macro- and microcirculation of adipose tissue. FEBS J 276:5738-5746, 2009. 9. Dixon JB. Lymphatic lipid transport: sewer or subway? Trends Endocrinol Metab 21:480-487, 2010. 10. Rutkowski JM, Markhus CE, Gyenge CC, Dermal collagen and lipid deposition correlate with tissue swelling and hydraulic conductivity in murine primary lymphedema. Am J Pathol 176:1122-1129, 2010. 11. Cockett AT, Goodwin WE. Chyluria: attempted surgical treatment by lymphatic venous anastomosis. J Urol 88:566-568, 1962. 12. O’Brien BM, Sykes P, Threlfall GN, et al. Microlymphaticovenous anastomoses for obstructive lymphedema. Plast Reconstr Surg 60:197-211, 1977. 13. O’Brien BM, Shafiroff BB. Microlymphaticovenous and resectional surgery in obstructive lympho­ edemas. World J Surg 3:3-15, 1979. 14. Degni M. New microsurgical technique of lymphatico-venous anastomosis for the treatment of lymphedema. Lymphology 14:61-63, 1981. 15. Degni M. New techniques of lymphatic-venous anastomosis for the treatment of lymphoedema. Cardiovasc Rivista Brasileira 10:175, 1974. 16. Clodius L, Piller NB, Casley-Smith JR. The problems of lymphatic microsurgery for lymphedema. Lymphology 14:69-76, 1981. 17. Clodius L. [Problems of microsurgery in lymphedema] Handchir Mikrochir Plast Chir 14:79-82, 1982. 18. Gloviczki P, Fisher J, Hollier LH, et al. Microsurgical lymphovenous anastomosis for the treatment of lymphedema: a critical review. J Vasc Surg 7:647-652, 1988. 19. Campisi C, Boccardo F. Role of microsurgery in the management of lymphoedema. Int Angiol 18:4751, 1999. 20. O’Brien BM. Replantation and reconstructive microvascular surgery. Part 2. Ann R Coll Surg Engl 58:171-182, 1976. 21. Campisi C, Boccardo F. Lymphedema and microsurgery. Microsurgery 22:74-80, 2002. 22. Ryan M, Campisi CC, Boccardo F, et al. Surgical treatment for lymphedema: optimal timing and optimal techniques. J Am Coll Surg 216:1221-1223, 2013. 23. Campisi C, Boccardo F, Campisi CS, et al. Surgery and translational lymphology: models for educational programs, research and clinical applications. J Siena Acad Sci 3:69-80, 2012. 24. Papendieck CM. The big angiodysplastic syndromes in pediatrics with the participation of the lymphatic system. Lymphology 31:390-392, 1998. 25. Campisi C, Accogli S, Boccardo F. Obesity and lymphedema in geriatrics: combined therapeutical approaches. BMC Geriatr 10(Suppl 1):A49, 2010. Available at http://www.biomedcentral.com/1471-2318/10/ S1/A49. 26. Campisi C, Bellini C, Campisi C, et al. Microsurgery for lymphedema: clinical research and long-term results. Microsurgery 30:256-260, 2010. 27. Campisi C, Davini D, Bellini C, et al. Lymphatic microsurgery for the treatment of lymphedema. Microsurgery 26:65-69, 2006. 28. Orletskiy AK, Timtschenko DO. [Use of devices for intermittent negative pressure therapy for treatment of athletes] NN Priorov Central Institute 1-11, 2009. 29. Campisi CC, Larcher L, Lavagno R, et al. Microsurgical primary prevention of lymphatic injuries following breast cancer treatment. Plast Reconstr Surg 130:749e-750e, 2012. 30. Dellachà A, Boccardo F, Zilla A, et al. Unexpected histopathological findings in peripheral lymphedema. Lymphology 33:62-64, 2000.

K23349_Neligan_33_Multi Lymph_r4_dc_0447-0462.indd 461

5/27/2015 9:38:16 AM

462

Part V  Current Treatment of Lymphedema

31. Campisi C, Eretta C, Pertrile D, et al. Microsurgery for the treatment of peripheral lymphedema: longterm outcome and future perspectives. Microsurgery 27:333-338, 2007. 32. Campisi C, Boccardo F, Tacchella M. Reconstructive microsurgery of lymphatic vessels: the personal methods of lymphatic-venous-lymphatic (LVL) interpositioned grafted shunt. Microsurgery 16:161166, 1995. 33. Boccardo F, Casabona F, De Cian F, et al. Lymphedema microsurgical preventive healing approach: a new technique for primary prevention of arm lymphedema after mastectomy. Ann Surg Oncol 16:703708, 2009. 34. Boccardo F, Casabona F, Friedman D, et al. Surgical prevention of arm lymphedema in breast cancer treatment. Ann Surg Oncol 18:2500-2505, 2011. 35. Boccardo F, Campisi CC, Molinari L, et al. Lymphatic complications in surgery: possibility of prevention and therapeutic options. Updates Surg 64:211-216, 2012. 36. Morotti M, Menada MV, Boccardo F, et al. Lymphedema microsurgical preventive healing approach for primary prevention of lower limb lymphedema after inguinofemoral lymphadenectomy for vulvar cancer. Int J Gynecol Cancer 23:769-774, 2013. 37. Boccardo F, De Cian F, Campisi CC, et al. Surgical prevention and treatment of lymphedema after lymph node dissection in patients with cutaneous melanoma. Lymphology 46:20-26, 2013. 38. Boccardo F, Dessalvi S, Campisi CC, et al. Microsurgery for groin lymphocele and lymphedema after oncologic surgery. Microsurgery 34:10-13, 2014. 39. Boccardo F, Casabona F, DeCian F, et al. Lymphatic microsurgical preventing healing approach (LYMPHA) for primary surgical prevention of breast cancer-related lymphedema: over 4 years followup. Microsurgery 34:421-424, 2014. 40. Rutkowski JM, Moya M, Johannes J, et al. Secondary lymphedema in the mouse tail: lymphatic hyperplasia, VEGF-C upregulation, and the protective role of MMP-9. Microvasc Res 72:161-171, 2006. 41. Thomas SN, Rutkowski JM, Pasquier M, et al. Impaired humoral immunity and tolerance in K14-VEGFR-3-Ig mice that lack dermal lymphatic drainage. J Immunol 189:2181-2190, 2012. 42. Olszewski WL, Engeset A, Romaniuk A, et al. Immune cells in the peripheral lymph and skin of patients with obstructive lymphedema. Lymphology 23:23-33, 1990. 43. Campisi C, Boccardo F, Campisi CC, et al. Reconstructive microsurgery for lymphedema: while the early bird catches the worm, the late riser still benefits. J Am Coll Surg 216:506-507, 2013. 44. Fujita T. Optimizing surgical treatment for lymphedema. J Am Coll Surg 216:169-170, 2013. 45. Hoffman JN, Fertmann JP, Baumeister RG, et al. Tumescent and dry liposuction of lower extremities: differences in lymph vessel injury. Plast Reconstr Surg 113:718-724, 2014. 46. Brorson H, Svensson H, Norrgren K, et al. Liposuction reduces arm lymphedema without significantly altering the already impaired lymph transport. Lymphology 31:156-172, 1998. 47. Lu Q, Delproposto Z, Hu A, et al. MR lymphography of lymphatic vessels in lower extremity with gynecologic oncology-related lymphedema. PloS One 7:e50319, 2012. 48. Brorson H. From lymph to fat: liposuction as a treatment for complete reduction of lymphedema. Int J Low Extrem Wounds 11:10-19, 2012. 49. Brorson H. From lymph to fat: complete reduction of lymphoedema. Phlebology 25(Suppl 1):52-63, 2010.

K23349_Neligan_33_Multi Lymph_r4_dc_0447-0462.indd 462

5/27/2015 9:38:17 AM

C hapter 34 Lymphatic Grafts Rüdiger G.H. Baumeister

K ey P oints • It has been shown experimentally in different groups that autologous lymphatic vessels are best suited for use as grafts. • One prerequisite for the use of these techniques is an isolated area of injury to the lymphatic transport system that is amenable to bypass from distal to proximal or peripheral to central with the use of a graft. • Reconstruction should be performed as soon as the source of lymphedema is identified.

Lym

• Different follow-up approaches have been used, such as volume measurements, nuclear medicine follow-up studies, and quality of life. • Lymphatic grafting for a patient with a locally interrupted lymphatic vascular system is a direct interventional procedure, with the main lymphatic collector grafts showing long-term patency.

The use of grafts is common in the field of vascular surgery as well as in plastic surgery. In vascular surgery, the bypassing of a regional vascular blockage with the use of vascular grafts is a basic principle with a long tradition, and it allows for the restoration of normal flow. As a result of the pathophysiology of lymphedema, the use of this technique is often indicated in patients with this condition. In Europe and the United States, most cases of lymphedema are the result of local damage to the lymph transport system, whereas globally filariasis-related lymphedemas are the most frequent ones. During the course of oncologic surgical procedures (especially those involving mammary carcinomas after the removal of lymph nodes in the axilla), diminished lymphatic transport may result, and lymphedema may develop. In addition, lymphedema may occur after surgical procedures in the pelvic and inguinal regions that involve the removal of lymph nodes. Severe trauma or surgery that interferes with “bottleneck” areas of the lymphatic transport system may be the cause of lymphedema distal to the area of destruction. There is also a specific group of patients

463

K23346_Neligan_34_Lymph Grafts_r4_dc_0463-0472.indd 463

5/26/2015 10:05:06 AM

464

Part V  Current Treatment of Lymphedema

with primary lymphedema who have locally deficient lymphatic systems in the inguinal and pelvic regions but normal peripheral lymph vessels. All of these local blockages may be overcome with the use of a vascular bypass procedure. It has been shown experimentally in different groups that autologous lymphatic vessels are best suited for use as grafts.

Graft Harvesting According to Kubik’s anatomic description, the ventromedial bundle at the inner aspect of the thigh contains up to 16 lymphatic collectors (Fig. 34-1). These spread out and form a network between the two critical lymphatic bottleneck regions of the lower extremity: the knee and the inguinal region. In this area of the medial thigh, lymphatic vessels can be taken out without touching the bottleneck areas for a length of up to approximately 30 cm. To ensure undisturbed lymphatic flow after the removal of the grafts, several specific steps should be taken: • The leg should be free of any preexisting lymphatic disturbances, and preoperative lymphoscintigraphy should confirm this. • During surgery, patent blue V dye is administered subdermally into the first and second web spaces. No sign of dermal backflow should be seen. • The lymphatic vessels will become stained after about 15 minutes.

Inguinal lymph nodes Great saphenous vein Harvested vessels

FIG. 34-1  The harvesting of two to three lymphatic vessel grafts from the ventromedial bundle of the thigh, which consists of up to 16 lymphatic vessels.

Ventromedial bundle

K23346_Neligan_34_Lymph Grafts_r4_dc_0463-0472.indd 464

5/26/2015 10:05:07 AM

465

Chapter 34  Lymphatic Grafts

• Stained lymphatic collectors should always be left in place and untouched during the preparation of the grafts, with only two to three collectors taken and the rest left undisturbed. • Lymphatic collectors often show distal branching. Therefore, when using two or three main collectors as grafts, several branches can also be incorporated so that a higher number of peripheral anastomoses can be performed. The open central ends of the transected vessels distal to the harvested grafts are carefully occluded with either sutures or coagulation to avoid leakage. The grafts can then be used in two different ways: they can be left attached to the inguinal nodes proximally and then “pedicled” to the scrotum or the opposite leg to treat penoscrotal and lower extremity lymphedema, or they can be used as free grafts for the treatment of the upper extremity. When the collectors are to be used as pedicled grafts, they are harvested, occluding them at the distal ends with long sutures; these sutures are used to pull the grafts to the penis, the scrotum, or the opposite leg. After anastomosing, the distal end is then ready for the influx of lymph from the edematous area, and the lymph will then flow in the correct direction back toward the inguinal nodes in the leg from which the collectors were harvested. Alternatively, the lymphatic collectors can be used as free grafts for the treatment of lymphedema of the upper extremity. For free grafting, the lymphatic vessels are transected distally. Centrally they are occluded with a suture with long threads. Once again, the sutures are used to pull the grafts into the desired position.

Grafting to Bypass an Axillary Blockage A graft procedure to bypass an axillary blockage is performed with the patient under general anesthesia. The patient’s arm is abducted, his or her neck is somewhat stretched, and the head is turned toward the opposite side (Fig. 34-2). Sternocleidomastoid muscle Internal jugular vein

FIG. 34-2  Connecting lymphatic collectors at the upper arm and the neck, with lymphatic vessel grafts bypassing the affected axilla.

Lymphatic collectors and nodes of the neck

Anastomosis near right lymphatic duct

Lymphatic vessel grafts

Lymphatic collectors of the arm

Area of lymph node and vessel dissection after breast-conserving surgery

K23346_Neligan_34_Lymph Grafts_r4_dc_0463-0472.indd 465

5/26/2015 10:05:07 AM

466

Part V  Current Treatment of Lymphedema

An oblique incision is created in the upper arm distal to the axilla. Because dye would be ineffectively transported within the edematous area and only a definitive area has to be checked, no staining is necessary. The biggest collectors are found above the fascia; they are gray and shiny. Care must be taken to distinguish lymphatic vessels from small veins, nerves, and fibrous cords. In a patient with long-standing lymphedema, the lymphatic vessels often have a thickened wall; however, they do not have oblique stripes like nerves do. The final decision about the quality of the vessel can only be made after transection. The lumen has to be demonstrable, and it should not contain any blood. A second incision is then created about 3 cm above the clavicle at the lateral border of the sternocleidomastoid muscle. The muscle is shifted medially, and the fatty tissue underneath is inspected toward the internal jugular vein. To facilitate the search for the tiny lymphatic vessels, a small amount of patent blue vital dye may be injected subdermally behind the patient’s ear before the incision is made. Small lymph nodes can also be found within the fatty tissue beneath the sternocleidomastoid muscle. Sometimes it is easier to connect the grafts to the lymph nodes rather than performing end-to-end or end-to-side anastomoses of the small, fragile lymphatic vessels. Between the incisions in the upper arm and the neck, a blunt tunnel is created, and an 18 Fr catheter is inserted. A suture is incorporated within the catheter to allow the grafts to be pulled through the tube. To avoid friction, the catheter is filled with Ringer’s lactate solution. After the grafts are pulled through, the cervical end is secured, and the catheter is removed distally. Attention is paid to both incisions. It is important to ensure that the length of the graft is sufficient for a tension-free anastomosis. Tension-free anastomosing means that no tension is applied to the vessel wall in the transverse direction; the fragile wall would be disrupted. Therefore the vessel is not turned, such as in the Cobbett technique. Of course, there should be no tension on the vessels in the longitudinal direction, but this is also clear in the anastomoses of other vessels. In the upper arm, end-to-end anastomoses are performed using a tension-free anastomosing technique. The first single stitch is placed opposite to the microsurgeon’s position. Subsequent sutures are started at the back wall. For small vessels, as few as three sutures may be sufficient; otherwise, sutures are inserted as necessary (Fig. 34-3). All of the sutures used are single stitches. Histologic findings in experimental studies have shown only minimal foreign body reactions around tiny vessels with absorbable sutures; this is in contrast with the significant foreign body reactions caused by nonabsorbable suture material. For this reason, only the smallest 10-0 absorbable polyglactin 910 suture is currently used. Although the nicely stained lymphatic vessels at the thigh can be harvested without magnification, the anastomosing procedure requires the highest magnification possible under the operating microscope. Searching for appropriate lymphatic vessels and discriminating among similar structures such as small nerves also requires magnification; however, medium-range magnification is often sufficient for these purposes.

K23346_Neligan_34_Lymph Grafts_r4_dc_0463-0472.indd 466

5/26/2015 10:05:07 AM

467

Chapter 34  Lymphatic Grafts

End-to-end anastomosis (draining lymph collector and lymphatic graft)

End-to-side anastomosis (lymphatic graft and lymphatic vessel)

End-to-lymph node capsule anastomosis (lymphatic graft and lymph node)

FIG. 34-3  Lymphatic-lymphatic and lymphatic to lymph node capsule anastomosis techniques.

Ventromedial bundle of the thigh Anastomosed short lymphatic vessel grafts Area of previous lymphatic vessel damage and fibrosis Scar from previous injury or surgery Excision of scar and new incision

FIG. 34-4  Short lymphatic vessel grafts bypass the interrupted lymphatic bottleneck at the knee.

Ventromedial bundle of the leg

Lymphatic collectors of the leg

Grafting Locally Blocked Lymphatic Pathways in the Periphery of the Extremities Surgical interventions or severe trauma at bottleneck areas of the lymphatic system, such as the medial aspect of the knee, may also result in peripheral lymphedema. To overcome these blockages, short lymphatic grafts can be anastomosed proximal and distal to the injured area (Fig. 34-4).

K23346_Neligan_34_Lymph Grafts_r4_dc_0463-0472.indd 467

5/26/2015 10:05:08 AM

468

Part V  Current Treatment of Lymphedema

Transposing the Grafts in Unilateral Leg Edema One of the patient’s legs is used when harvesting the graft. The grafting procedure is the same as it is for free grafting in the case of arm edema, except for the proximal transection. The proximal attachment to the inguinal lymph nodes remains intact so that there is no proximal transection. On the edematous leg, if there is a blockage in the pelvic or inguinal area, an incision is made distal to the inguinal region. This may occur after urologic or gynecologic procedures in which the lymph nodes are removed at or above the scar of a previous intervention. Lymphatic vessels are identified for anastomosis to the graft (Fig. 34-5). A tunnel is created between the proximal end of the dissection at the harvesting site and the incision on the opposite side, and an 18 Fr catheter is inserted. The distal end of the graft is transfixed with a long suture and pulled through the catheter, which is lubricated to allow for the easy passage of the graft. The catheter is removed from the incision site of the affected limb, and the anastomosis is performed.

Inguinal lymph nodes in healthy leg Anastomosed long lymphatic vessel grafts Lymphatic collectors of the leg

Leg edema

FIG. 34-5  Lymphatic collectors remaining attached at the inguinal lymph nodes in the healthy leg are transposed to the edematous side and anastomosed with main lymphatic collectors of the affected leg.

K23346_Neligan_34_Lymph Grafts_r4_dc_0463-0472.indd 468

5/26/2015 10:05:08 AM

Chapter 34  Lymphatic Grafts

Fibrosed inguinal lymph nodes and vessels

Penile and scrotal edema

469

Healthy lateral inguinal lymph nodes

Anastomosed short lymphatic collectors from the thigh

Lymphatic collectors of the penis and scrotum

FIG. 34-6  Short lymphatic collectors remaining attached at the inguinal lymph nodes in the healthy leg are transposed to the penile and scrotal region and anastomosed with patent lymphatic collectors.

Transposing the Grafts in Penile or Scrotal Edema A healthy leg is used for harvesting, and the procedure is similar to that used for unilateral leg edema. However, lymphatic vessels that drain to the lateral inguinal lymph nodes are preferred for harvesting, because the medially located lymph nodes that drain the penis and the scrotum may be affected. The search for lymphatic vessels that are appropriate for harvesting begins at the root of the penis or the scrotum. The distance to the harvesting site is short, so short grafts are sufficient. The procedure of transposition is the same as for unilateral leg edema (Fig. 34-6).

Indications One prerequisite for the use of these techniques is an isolated area of injury to the lymphatic transport system that is amenable to bypass from either distal to proximal or peripheral to central with the use of a graft. Long-standing lymphedema damages the lymphatic vessels and leads to significant tissue alterations. Therefore reconstruction should be performed as soon as the source of lymphedema is identified. Sufficient time is allowed to elapse to ensure that any transient edema dissipates, and a short trial of conservative treatment is reasonable to allow the patient the opportunity to solve the problem without further treatment, thus avoiding surgery. A minimum of 6 months should be sufficient.

K23346_Neligan_34_Lymph Grafts_r4_dc_0463-0472.indd 469

5/26/2015 10:05:09 AM

470

Part V  Current Treatment of Lymphedema

Primary lymphedemas caused only by a local blockage are rare. These conditions can also be treated with grafting after a thorough preoperative evaluation by MR lymphography to reveal possible further alterations of the lymphatic system. An appropriate harvesting site must exist so that grafts of sufficient length can be used with minimum risk. The chosen area should also be checked with lymphoscintigraphy.

Confirming the Results Different approaches have been used for follow-up, including volume measurements, qualityof-life improvements, calculating the lymphatic transport by lymphoscintigraphy, and the documentation of complications. The basic follow-up procedure involves measuring the volume of the extremities; this yields more accurate data than just measuring their circumferences. The volumes are calculated using Kuhnke’s method, which involves measuring the extremity’s circumference every 4 cm and then adding the different volumes to obtain the total volume. These measurements are obtained before and after surgery at various intervals. It has previously been demonstrated that significant volume reduction was seen after 10 years for arm edema and after 4 years for leg edema. The mean volume reduction was about two thirds of the original preoperative volume. Even more important is the impact of this reduction on quality of life. In an assessment study of 212 patients by Springer et al, significant improvements in physiologic and psychological aspects occurred after surgical treatment. This was particularly noticeable for patients with arm edema after removal of the burden of conservative treatment, especially the diminished need to wear elastic stockings and undergo time-consuming manual lymphatic treatment. The evaluation of possible complications is also important during follow-up. To date, we have seen one lymph cyst at the harvesting site in one patient; this was treated by puncture. Another patient developed a swelling of the lower leg as a result of a venous thrombosis, and two others in an early series developed erysipelas postoperatively. No additional postoperative cases of erysipelas have been seen since we introduced the use of perioperative cephalosporin. In one patient, we treated a venous bleed that developed in the connecting tunnel made between the two incisions. Proving, measuring, and comparing the results obtained by different investigators is another important approach. In bypass procedures, questions arise about the patency of the grafts. Of the various methods used to detect patency, direct lymphography with an oily contrast medium is obsolete. Indirect lymphography with a water-soluble contrast medium stains mostly blood vessels, and only for a short distance. Although MRI lymphography is quite expensive, it is also highly sophisticated and effective. With the use of a water-soluble contrast medium and MRI lymphography, patent grafts have been demonstrated more than 10 years after grafting.

K23346_Neligan_34_Lymph Grafts_r4_dc_0463-0472.indd 470

5/26/2015 10:05:09 AM

Chapter 34  Lymphatic Grafts

471

Patency may also be proved indirectly with lymphoscintigraphy. This is especially useful for patients with unilateral lymphedema. In these cases, lymphatic grafts are transposed from one side to the other. By injecting the radiopharmaceutical only into the affected side, activity can be found within the lymph nodes of the contralateral side, thereby indicating lymph transport through the grafts to the healthy side. Often such transport may be seen more clearly after activity by exactly following the routes of the grafts. In addition to showing the patency of lymphatic grafts, lymphoscintigraphy is mainly used to check the functional aspects of lymphatic transport. With the help of the lymphatic transport index, a semiquantitative measurement of lymphatic outflow at an extremity is possible. Significant improvement can be demonstrated in both the upper and lower extremities by comparing the preoperative and postoperative transport indexes. Within a group of patients with imageable activity along the grafts, it is also possible to achieve normalization of the transport. Lymphatic grafting for a patient with a locally interrupted lymphatic vascular system is a direct interventional procedure, with the main lymphatic collectors showing long-term patency. This provides the patient with clear functional and clinical benefits as well as significant improvements in quality of life.

C linical P earls • The established procedure in vascular surgery of overcoming a local interruption is also possible within the lymphatic vascular system. • Normalization of diminished lymph flow is the key point for causal lymphedema treatment. • When lymph flow is restored first, additional treatment may be avoided after fat reduction with the use of liposuction in advanced stages of lymphedema.

BIBLIOGRAPHY Baumeister RG, Frick A. [The microsurgical lymph vessel transplantation] Handchir Mikrochir Plast Chir 35:202-209, 2003. Baumeister RG, Seifert J, Wiebecke B, et al. Experimental basis and first application of clinical lymph vessel transplantation of secondary lymphedema. World J Surg 5:401-407, 1981. Baumeister RG, Seifert J, Wiebecke B. Transplantation of lymph vessels on rats as well as a first therapeutic application on the experimental lymphedema of the dog. Eur Surg Res 12(Suppl 2):7, 1980. Baumeister RG, Siuda S. Treatment of lymphedemas by microsurgical lymphatic grafting: what is proved? Plast Reconstr Surg 85:64-74; discussion 75-76, 1990. Földi M. [The physiology of the lymphatic vessels] Angiologica 8:212-235, 1971. Kinmonth JB. The Lymphatics, ed 2. London: Edward Arnold, 1982.

K23346_Neligan_34_Lymph Grafts_r4_dc_0463-0472.indd 471

5/26/2015 10:05:09 AM

472

Part V  Current Treatment of Lymphedema

Kleinhans E, Baumeister RG, Hahn D, et al. Evaluation of transport kinetics in lymphoscintigraphy: followup study in patients with transplanted lymphatic vessels. Eur J Nucl Med 10:349-352, 1985. Kubik S. [Clinical anatomy of the lymphatic system] Verh Anat Ges 69:109-116, 1975. Kuhnke S. [Volume determination from circumference measurements] Folia Angiol 24:228, 1976. Springer S, Koller R, Baumeister RG, et al. Changes in quality of life of patients with lymphedema after lymphatic vessel transplantation. Lymphology 44:65-71, 2011. Weiss M, Baumeister RGH, Hahn K. [Planning and monitoring of autologous lymph vessel transplantation by means of nuclear medicine lymphoscintigraphy] Handchir Mikrochir Plast Chir 35:210-215, 2003. Yuwono HS, Klopper PJ. Comparison of lymphatic and venous interpositional autografts in experimental microsurgery of the canine lymphatics. Plast Reconstr Surg 85:752-757; discussion 758-759, 1990.

K23346_Neligan_34_Lymph Grafts_r4_dc_0463-0472.indd 472

5/26/2015 10:05:09 AM

C hapter 35 Lymphaticovenular Anastomosis Isao Koshima, Mitsunobu Harima

K ey P oints • Excision of a lymph node in the extremities causes dilation of the peripheral lymphatic vessels and degeneration of smooth muscle cells within a short period. • Although smooth muscle cells regenerate, they remain small unless full restoration of lymph flow is achieved. • Combination therapy that consists of the reconstruction of lymph reflux function by microscopic lymphatico­ venular anastomosis and compression is an effective treatment for this condition. • Although anastomosis is effective as a preventive and early stage application, it is also effective for patients with chronic edema (lymphedema). • In the future, a method will be developed to reconstruct the reflux function based on vascularized lymphatic implantation, in addition to the promotion of preventive anastomosis.

Lym Both conservative therapy and surgical procedures (for example, tissue ablation and classic lymph– vascular conduction surgery) have been performed to treat lymphedema. Conservative therapy is directed at slowing the progression of edema, and surgical interventions attempt radical treatment by redirecting the flow of retained lymph back to the venous system by an artificially prepared bypass. However, it is difficult to significantly improve prolonged edema with either of these methods.1,2 With the recent advent of the supermicrosurgery technique (vascular anastomoses 0.5 to 0.8 mm in diameter) in plastic and reconstructive surgery, it has become possible to create an anastomosis with extremely tiny blood vessels.3-6 Based on the microscopic anastomosis technique, a new method of lymphaticovenular anastomosis (LVA) has been developed that surpasses the performances achieved by conventional macroscopic lymph–vascular vein conduction1,2,7 or LVA.8-10 This chapter is adapted from Koshima I, Narushima M, Mihara M, et al. Treatments for leg lymphedema. J Jpn Soc Gynecol Oncol 27:10-18, 2009.

473

K23349_Neligan_35_Lymphatico Anastom_r3_dc_0473-0486.indd 473

5/27/2015 12:42:47 PM

474

Part V  Current Treatment of Lymphedema

Currently patients undergoing surgical treatment in combination with anastomosis and compression therapy have been followed for more than a decade.3-8 As a result, the conventional notion that “lymphedema is incurable” is shifting to a concept that the disease can be cured by early or preventive anastomosis or by combination therapy based on anastomosis and compression.

The Fate of Lymphatic Vessels With Lymphedema Lymphedema develops in patients with congenital hypoplasia when there are few collateral pathways in the lymphatic system to drain an area. For this reason, the removal of even a few lymph nodes on the medial side of an extremity may cause rapid dilation and failure in the related and nearby lymphatic vessels (if there are any). As a result, the smooth muscle cells of the remaining and central lymphatics degenerate soon after the removal of the lymph nodes. Later, the smooth muscle cells may regenerate but often remain in a hypertrophied state.3 Thus it is impossible to restore strong systolic contractile function of these remaining lymphatic vessels, resulting in a continued reflux disorder. In patients with repeated lymphangitis during this time, occlusion and destruction of lymphatic vessels occur. Furthermore, there is growth of collagenous fibers formed by activated fibroblast cells between the subcutaneous fat tissues and the dermis. However, based on our knowledge of all biopsy findings in lymphatic vessels at the time of surgery of our clinical cases, the morphologic evidence of degeneration and regeneration of smooth muscle cells differs, depending on individual cases and sites within the extremities (Fig. 35-1).

A

B

FIG. 35-1  Lymphatic vessel biopsy find­ ings of a patient with edema of the lower limb. A, In this optical microscope image, lymphatic vessels in the central part of the lower thigh are significantly dilated, whereas the thickness of the media is kept properly. B, Electronic microscopy shows a greater growth of collagenous fibers in the media, with many large-sized smooth muscle cells. In this edema case, the reflux function of lymphatic vessels may have been preserved relatively well despite the extremely pro­ longed term.

K23349_Neligan_35_Lymphatico Anastom_r3_dc_0473-0486.indd 474

5/27/2015 12:42:47 PM

Chapter 35  Lymphaticovenular Anastomosis

475

The duration from the onset of edema has no correlation with the degree of regeneration.3 However, the degree of regeneration of the smooth muscle cells is to some extent correlated with the degree of postoperative reduction in edema and cellulitis. Moreover, even if lymphatic vessels are destroyed throughout the area, lymphatic vessel function in the distal extremity is frequently preserved. As a result, partial improvement is often achieved in many patients with bypass surgery only in the peripheral parts. Also, because some reduction in edema is observed in the femoral region by anastomosis at the ankle joint in some patients, it may be true that valvular insufficiency occurs in the lymphatic vessels in edema patients, causing lymphatic return.

Assessment of Lymphatic Functional Status Conventional methods, such as lymphoscintigraphy, can indicate the location of lymph collectors but cannot provide information about the functional state or specific areas of occlusion. As a test for lymph collector functionality, the recently developed fluorescence lymphography shows promise. By applying infrared rays to subcutaneously injected indocyanine green (ICG), it is possible to confirm that the ICG is rapidly flowing back through lymphatic vessels while showing fluorescence in the normal extremities. In the edematous region, it is possible to easily confirm the degree of reflux under direct vision11 (Fig. 35-2).

A

B

FIG. 35-2  Lymphatic vessel fluorescent imaging. A, The imaging result of both lymphatic vessels of the lower limbs have been traced on a patient with edema of both lower limbs. Even in patients with mild edema, imaging is often performed not at the proximal side but only at the distal side. B, Applying infrared rays to ICG after intradermal injection, ICG is observed flowing back through lymphatic vessels.

K23349_Neligan_35_Lymphatico Anastom_r3_dc_0473-0486.indd 475

5/27/2015 12:42:48 PM

476

Part V  Current Treatment of Lymphedema

In mild lymphedema, whereas fluorescently stained lymphatic vessels are generally not observed in the proximal or medial part of the limb, a few vessels may be recognized in the more peripheral and distal areas. With unilateral lymphedema of the lower limb, these vessels do not fluoresce, even in unaffected lower limbs in many cases. This suggests that the reflux function of the lymphatic vessels may have been significantly lost in patients with lymphedema of the lower limb, including that of the unaffected side at the time edema developed.

Treatment Methods The major treatment method relevant to LVAs is compression therapy with elastic stockings.

Long-Stretch Elastic Stockings Stockings worn to prevent edema formation work by applying external pressure to enhance tissue pressure. This is a core, indispensable therapy, even for mild edema, and can be used daily from the early stages of edema. Patients are instructed to wear them without exception during the day when standing. As the stockings loosen (usually after about 6 months because of frequent and repeated washing), they should be replaced with a new pair. These are often adapted to fit individuals by adding a fastener (zipper) or an elasticized band, as required. For patients who require the stocking larger than standard size or have irregular-shaped limbs, a custom-made stocking is ordered. With the Földi method (which uses compression concomitant with intensive massage therapy based on hospitalization), surgical treatment is a contraindication.12

Lymphaticovenular Anastomosis LVA is a permanent solution for the lymphedema, rerouting lymph back to the venous system through a bypass prepared for it with no need for compression therapy. We have performed 20 to 30 LVAs, guided by supermicrosurgical magnification of the site. A fine needle with a 50-micron (1∕20 mm) suture (Fig. 35-3) is used to link the vessels. Our operative method is different from the conventional lymphaticovenous anastomosis13 in that the cutaneous veins to be refluxed are connected with tiny venules immediately beneath the dermis or in the shallow part of the fat layer in an end-to-end anastomosis.3-6 In principle, the anastomosis is performed after the use of conservative therapy (mainly by outpatient treatment) based on intensive continued compression for at least 6 months before surgery. The compression therapy is continued even after surgery (combination therapy). With recently developed fluorescence lymphography, it is possible to identify lymphatic vessels with normal reflux function by observing their presence under the skin. With the use of a local anesthetic during surgery, it is possible to readily expose those lymphatic vessels with good functionality with a small incision.11 As a result, this operative method, which should be performed only by experienced surgeons, is being increasingly adopted by facilities worldwide.

K23349_Neligan_35_Lymphatico Anastom_r3_dc_0473-0486.indd 476

5/27/2015 12:42:48 PM

Chapter 35  Lymphaticovenular Anastomosis

477

A

B

FIG. 35-3  LVA during surgery. A, In a lower limb edema case, reconstructive surgery is performed by three teams simultane­ ously using three microscopes. The procedure is performed with the use of a local anesthetic. B, Microscopic LVA. Magnified by the microscope about 303, lymphatic vessels and venules of 0.5 mm in diameter are sutured by six to eight stitches using a needle 50 microns in diameter. Reflux of lymph fluid from lymphatic vessels into venules is observed immediately after surgery.

History of Lymphaticovenular Anastomosis The first reported experimental lymphaticovenous anastomosis was performed by Laine in 1963.12 Yamada14 first performed clinical application of lymphaticovenous anastomosis for obstructive lymphedema in 1967. Degni7 described end-to-side anastomoses between a saphenous vein and surrounding lymphatic vessels with a single stitch in the inguinal region. O’Brien8 described the same approach to treat lymphedema referring to Yamada’s procedure. However, these procedures were not widely accepted, because outcomes of the treatments were poor. Nevertheless, the achievements of these pioneers laid the foundations for the subsequent development of lymphedema surgery. The introduction of supermicrosurgical technique in the 1980s was the critical development in reconstructive microsurgery. This technique was used to provide successful fingertip replantation and contributed to the development of the deep inferior epigastric perforator (DIEP) flap, the initial perforator flap.15,16 Koshima3-6 applied supermicrosurgical technique for LVA to manage lymphedema patients in the 1990s, and further improvement of edema more than before was obtained. This technique made possible the precise anastomosis of vessels smaller than 0.5 mm in diameter. The development of supermicrosurgical instruments and suture materials helped to ensure more accurate surgical approach for lymphedema. Based on our experience, we recommend the use of multiple surgical microscopes at the same time to perform multiple LVAs quickly and to increase the number of anastomosis (see Fig. 35-3). We also recommend the use of elastic stockings after surgery for at least half a year.

K23349_Neligan_35_Lymphatico Anastom_r3_dc_0473-0486.indd 477

5/27/2015 12:42:49 PM

478

Part V  Current Treatment of Lymphedema

LVAs have proved to be effective, especially in patients in the earlier stages of lymphedema (stage 1 or 2), since here the mechanism of degeneration of lymphatic channel smooth muscle cells is well understood.3 However, LVA only is not fully effective for severe and advanced edema. For those cases, we now use combined surgical treatments with multiple LVAs and functional lymphatic transfer.

Surgical Technique LVA requires highly sophisticated microsurgical technique called supermicrosurgery, and no training model specific to LVA has been developed. In our center, before surgery, ICG is injected subcutaneously in the peripheral extremities. Using an infrared lamp, it is possible to confirm that the ICG is flowing up through the lymphatic vessels. At this stage the location and paths of the lymph collectors should be traced using a marker pen. In many cases, lymphatic vessels are only identified on the peripheral side of the extremities. After the affected extremities are disinfected, an incision line 3 cm in length is made over the lymphatic vessels identified by the ICG. Where lymphatic vessels are not identified, incision lines should be placed on 5 to 10 sites on the medial and lateral side of the affected extremities. The reason for this is that many lymphatic vessels and main cutaneous veins are located at this site and smaller veins which are branches of them are easily found. While it is difficult to find lymphatic vessels in the upper part of the arms and in the femoral regions, they are often found in forearms and lower thighs even if they were not identified by ICG. Further, in the case of unilateral lower limb edema, a preventive LVA should be planned at the ankle joint on the unaffected side as edema can develop on the unaffected side. Vascularization is carried out by Esmarch bandage and tourniquet at the proximal part of an affected extremity with the patient under general anesthesia, whereas we usually perform surgeries with local anesthesia using 1% lidocaine (xylocaine E). After the skin incision has been made, venules that lie immediately beneath the dermis or in the shallow part of the fat layer (with a diameter of 0.6 to 1.0 mm) and the adjacent lymphatic vessels are exposed and separated. Although subcutaneous injection of dye (patent blue is most effective) is thought to be useful for identifying lymphatic vessels, and normal lymphatic vessels are densely stained by this dye, it is often inappropriate for vessels in patients with advanced lymphedema: it has lower stainability because of reduced transport of the dye. If both lymphatic vessels and veins exist in the same incision site, they should be marked by a vessel tape or 3-0 nylon suture. Of 10 incision sites usually made, lymphatic vessels are generally observed only at about half of them or occasionally are not found at all. If lymphatic vessels are cut off, reflux of lymph is preferable at both cutting distal and proximal sites in the distal lower thigh but poor at the femoral part and groin, exhibiting hardening of the lymphatics. If reflux is in an acceptable state, some improvement in edema is often obtained after surgery by just one anastomosis. When venous flow direction is confirmed on the central and peripheral side by a patency test, anastomosis is carried out so that lymph flows in an antegrade direction to the veins. As end-to-end or end-to-side anastomosis to connect a lymphatic vessel with a vein of the same diameter is carried out under the microscope; we create them with six to eight stitches of inter-

K23349_Neligan_35_Lymphatico Anastom_r3_dc_0473-0486.indd 478

5/27/2015 12:42:49 PM

Chapter 35  Lymphaticovenular Anastomosis

479

rupted suture using 11-0 or 12-0 nylon (both require a needle with a diameter of 30 to 50 microns or smaller) and a 50-micron needle. While clipping the venous side (the clip is often unnecessary if a valve exists proximally) and allowing lymph to leak from the lymphatic vessel, the anastomosis is carried out under maximum magnification (303). To avoid rupture of the lymphatic vessel wall by irregular hand movements when passing a needle through the vessel, the surgeon should hold his or her breath. If two veins are secured, the anastomotic branch should be on the proximal side of the lymphatic vessel (since lymph flow is often reversed). Influx of lymph into the veins is seen after the anastomosis is completed. In some cases, a cellulitis-like symptom may appear immediately after surgery as a result of reflux of blood into the lymphatic vessels. This can occur when the vein to be anastomosed has a diameter of 1.5 mm or larger, or sometimes just by venous reflux. To prevent postoperative occlusion at the anastomotic site, a continuous drip of prostaglandin E1 is given for about 5 days, and an oral form is administered for several weeks. Because the incisions are not deep, there is very little postoperative pain, and the patient can go home the day of surgery. In both upper and lower extremities cases, walking is permitted immediately after surgery, and continuous compression therapy is resumed 3 weeks after the surgery. In cases in which there were large reductions (especially in the upper extremities), compression is experimentally and intermittently released 6 months after surgery, and the conservative therapy is gradually suspended (see Fig. 35-3).

Comparison of Compression Therapy and Combination Therapy Of 35 patients with unilateral or bilateral lymphedema in the lower limbs with an average duration of 5.2 years, 12 (34%) underwent conservative therapy mainly based on compression by outpatient visits. For this group, the average excessive circumference of 7.1 cm of the lower thigh (measured 10 cm proximal to the midpoint of the patella) before surgery decreased by an average of 0.6 cm after 1.5 years of conservative therapy. A large decrease in the circumference of more than 4 cm was observed in two cases (6%). On the other hand, surgery was performed on 16 patients (46% of the total) whose lymphedema never resolved from conservative therapy at other facilities or in our department. Preoperative excessive circumference was 9.8 cm on average in these patients, with an average of 8.9 years since its onset. An average decrease of 2.7 cm was achieved 3.3 years on average after the surgery. A decrease in the circumference by 4 cm or larger was observed in eight cases (50%).3 These results indicate that a more favorable effect is obtained not by compression therapy alone but in combination with the creation of anastomoses.

Case Variations Lymphaticovenular Anastomosis at an Early Stage After Onset A significant effect is expected for edema before onset or at an early stage after onset with our operative method. During a period before onset of edema or at an early stage when the smooth muscle cells of the lymphatics still function, lymphedema is prevented by just one anastomosis in some cases. In our recent summary (unpublished date), one to three anastomoses (average 1.4)

K23349_Neligan_35_Lymphatico Anastom_r3_dc_0473-0486.indd 479

5/27/2015 12:42:49 PM

480

Part V  Current Treatment of Lymphedema

were performed in 35 patients with early stage edema in the lower limb within 9 months (average 2.3 months) after onset. Follow-up was performed for 6 years (average 31.7 months) after surgery. The anastomoses were effective in 88.5% of cases; in 11.5% no improvement was observed.

Patients Requiring No Postoperative Compression Therapy Restoration of lymphatic vessel reflux function is observed in some cases just by simple compression for several months or years after surgery. Of about 200 cases of upper and lower limb edema for which anastomosis has been carried out, preoperatively observed progression has been suspended without applying compression after surgery in four cases of upper limb and six cases of lower limb. As for the breakdown, with the term of follow-up after surgery ranging from two months to 15 years, both mild and severe cases were observed within a year since onset of edema in three cases of upper limb, and restoration was observed in a severe case after 11 years since the onset. In lower limb cases, it was also effective for both mild and severe cases within a year since the onset, showing effectiveness also in mild cases after 11 years from the onset. From these results above, it is believed that regeneration of smooth muscle in lymphatic vessels is different by individuals depending on the site and symptoms and that, in such cases that require no compression after surgery, regenerated smooth muscle is further differentiated by anastomosis resulting in restoration of lymph reflux function.

Patients With Lymphedema of Long Duration In general, in patients whose lymphedema has been present more than 10 years, it is rare that an anastomosis will produce a satisfactory outcome, because significant subcutaneous fibrosis is often observed under the operational microscope. However, even in these cases a reasonable outcome can be achieved by employing long-term postoperative compression therapy. Combination therapy still generally remains more effective for upper limb intervention compared with conservative therapy alone. In cases of lower limb lymphedema, combination therapy is also effective for about half of the cases, even if reduction of the circumference of the limb was not obtained. An important observation is that the incidence of cellulitis was remarkably reduced in cases in which there had been frequent episodes before the intervention.3-6 Even in these patients, when a functional lymphatic vessel could be anastomosed, a significant decrease in volume was usually achieved compared with that before surgery. The 42-year-old woman in Fig. 35-4 had early surgery after the onset of edema. Severe lymphedema then appeared in both lower limbs immediately after excision of uterine cancer (Fig. 35-4, A). Anastomosis was carried out earlier because of rapid exacerbation despite the fact that compression therapy with stockings was instituted after surgery. One anastomosis was carried out for both knees on the medial side (Fig. 35-4, C and D) under local anesthesia 3 or 4 months after onset of the edema. Two years after surgery (Fig. 35-4, B), her edema has been remarkably improved by using compression therapy concomitantly (Fig. 35-4, E).

K23349_Neligan_35_Lymphatico Anastom_r3_dc_0473-0486.indd 480

5/27/2015 12:42:49 PM

481

Chapter 35  Lymphaticovenular Anastomosis

A

B

C

E

D

61 Right thigh Left thigh 56

51

46

Pre­ operative

3 mo post­ operatively

11 mo

1 yr 4 mo

2 yr

FIG. 35-4  A, This 42-year-old woman developed bilateral lymphedema after surgery for uterine cancer. B, Two years after surgery. C, Medial side of right knee. D, Medial side of left knee. E, Change in circumference of both lower thighs (10 cm be­ low the knees) 2 years after surgery. Although an increase in circumference was observed before surgery despite applying compression, it continued to decrease immediately after surgery.

K23349_Neligan_35_Lymphatico Anastom_r3_dc_0473-0486.indd 481

5/27/2015 12:42:50 PM

482

Part V  Current Treatment of Lymphedema

This woman, also 42 years old (Fig. 35-5, A), developed idiopathic bilateral edema of the lower thighs. Despite several surgeries performed at other medical facilities, including the Thompson method, no improvement was observed, and the lymphedema had a tendency to exacerbate. Cellulitis occurred frequently despite continuous compression therapy. One anastomosis was carried out for each of five incision sites at the medial side of right lower thigh (Fig. 35-5, B) (that is, five anastomoses in total), 24 years after the initial onset of edema. Because a lymphatic vessel was not identified in the left lower limb, an island adipofascial flap from the left inguinal region was transferred to a site under the skin in the anterior thigh (Fig. 35-5, C and D). With a significant improvement in circumference observed in the right and left lower thighs (a reduction of 13.5 and 11 cm, respectively) 8 years after surgery (Fig. 35-5, E), she can once again work at her daily job (sewing on a sewing machine) with almost no development of cellulitis. The circumference of her lower thighs has been reduced on both sides over time just by simple compression after surgery (Fig. 35-6).

A

B

C

D

V

V

L L

E

FIG. 35-5  A, Bilateral lower thigh edema. B, Incision sites at the medial side of right lower thigh. C, Lymphatic vessel (L) and venule (V) anastomosed; 11-0 nylon suture was used with an 80-micron needle . D, After anastomosis. E, The patient is seen at follow-up 8 years after surgery.

K23349_Neligan_35_Lymphatico Anastom_r3_dc_0473-0486.indd 482

5/27/2015 12:42:51 PM

483

Chapter 35  Lymphaticovenular Anastomosis

45 Left knee Right knee 40

35 Pre­ operative

4 mo post­ operatively

1 yr

2 yr

5 yr

7 yr

8 yr

FIG. 35-6  Change in circumference of both knees up to 8 years after surgery.

Contraindications to Anastomosis and Treatment of Recurrences Anastomosis is not appropriate in about 10% of the cases with early stage lymphedema (stage 1 or 2). These are generally patients with lower limb lymphedema for whom compression therapy after surgery has been halted, and severe fibrosis is observed, with surgical findings of lymphatic vessel sclerosis or occlusion. In these cases, sufficient entry of lymph into the venous system may not be obtained, even if an anastomosis was completed. Even in some patients who have achieved improvement postoperatively, lymphedema may continue to worsen even with the continual application of compression. As a countermeasure for this worsening, it is sometimes necessary to improve the flow of lymph into the venous system by establishing additional paths for it by creating additional anastomoses, while continuing the compression therapy.

Frequency and Prevention of Bilateral Lower Limb Lymphedema In our 356 lower limb lymphedema cases, a significantly large number of patients had lymphedema after treatment for uterine cancer or for malignancy of urinary structures (87%). There were a small number of idiopathic cases. Of the idiopathic cases, 44% were bilateral. It was interesting to note that unilateral edema may become bilateral at about 9 years after onset. Twenty-six percent of the secondary lymphedema cases were bilateral, and about 26% of these became bilateral immediately after surgery, showing a predominant tendency to shift from unilateral to bilateral type by about 69%. On average, it is common for lymphedema to appear unilaterally 41∕2 years after surgery and then to shift contralaterally to bilateral type at about 7 years after surgery. The overall percentage of patients whose lymphedema transitioned from unilateral to bilateral was 26%.

K23349_Neligan_35_Lymphatico Anastom_r3_dc_0473-0486.indd 483

5/27/2015 12:42:51 PM

484

Part V  Current Treatment of Lymphedema

During preoperative compression therapy, we had one patient with unilateral lower limb edema that had rapidly shifted to the bilateral type and had worsened. This underscores the fact that even with a unilateral case, it is not always possible to prevent the unaffected contralateral limb from developing edema. However, in a long-term follow-up after preventive or early stage anastomosis—which we have been advocating for more than a decade—its effectiveness in this case has been proved.5,6

Genital Lymphedema (Fistula) The QOL of genital lymphedema patients is significantly impaired, since the progressive tendency of the condition often leads to the development of bilateral leg lymphedema. Genital lymphedema occurs from cancer treatment in the genital region. There may be alteration of sexual and urinary function, and it is difficult to use compression therapy. To attempt to remediate this, we have performed LVA in 13 cases of refractory genital refractory edema and stoma (age range 14 to 80; 8 males, 5 females) with the patient under local anesthesia. Bilateral lower limb edema was an existing complication in 10 patients; 3 cases were primary and 10 were secondary. The duration from the onset of edema (lymphorrhea) ranged from 2 months to 12 years (average 4 years and 4 months); the number of lymphatic vessels for which anastomosis was performed was 1 to 5 vessels (average 2.6); and postoperative follow-up ranged from 2 months to 2 years and 3 months (average 13 months). Compression therapy was performed before and after surgery for the patients who had preexisting lower limb edema. In these cases compression therapy produced no effect. Intraoperative flow of lymph into veins was achieved in multiple anastomoses (67.6% of all 34 anastomoses), including those for the lower limbs. It was possible to secure at least one favorable anastomosis in most patients, resulting in remarkable reduction of edema after surgery and even improvements in prior lymphorrhea. One patient had lymphorrhea for which five towels per day were required to absorb the lymph drainage, and this flow was halted. In cases of genital edema (lymphorrhea) of long duration, LVA is likely to reduce symptoms. The explanation for this may be that lymph reflux (into the venous system) is preserved to a large extent, even in chronic cases of genital edema.

Therapeutic Strategy in the Future: The Need for More Effective Surgical Therapy In our experience, preventive anastomosis before or immediately after the onset of lymphedema may reduce the chance of its future development or exacerbation.6,7 One study reported that the frequency of edema occurrence was reduced by performing omental transposition concomitant with uterine cancer surgery (Dr. Keiichi Fujiwara, personal communication, 1998). Similarly, we must show that immediate anastomosis performed simultaneous with lymph node extraction can effectively reduce the risk of subsequent edema.

K23349_Neligan_35_Lymphatico Anastom_r3_dc_0473-0486.indd 484

5/27/2015 12:42:52 PM

Chapter 35  Lymphaticovenular Anastomosis

485

We have recently worked on a method to reroute lymph into the venous system by transplantation of lymphatic vessels vascularized with normal reflux function from the unaffected side in patients with LVA-resistant, progressive, severe lymphedema. Lymphedema can be totally prevented by combined modality therapy based on a team approach, including plastic surgery, along with the surgery associated with the various cancers and disorders handled by the breast surgical, obstetrics and gynecology, and urology teams.

Conclusion Sophisticated supermicrosurgical techniques have been developed that make it possible to perform vascular anastomosis of smaller vessels, and it has proved safe and effective to perform precise anastomoses between lymphatic vessels and venules. In the future it may become important to proactively select a surgical therapy that corresponds to the degree of functional disorder of the lymphatic system, which could be determined by genetic screening.

C linical P earls • The goal of surgical treatments for lymphedema is complete recovery or elimination of the need for physical therapy and compression therapy. • Smooth muscle degeneration same as in skeletal muscle denervation is the key mechnism in the progression of edema. • To perform LVA, the surgeon must have supermicrosurgical training and be proficient in the use of a fine needle with a 50-micron (1∕20 mm) suture. • Prophylactic LVA to preserve smooth muscle cells is a sound, effective strategy for all lymphedema. • Early-stage edema in the upper arm can be completely cured with LVA. • Treatment of late-stage or advanced edema should be performed with combined surgical treatments with LVA and functional lymphatic transfer. • Liposuction and partial resection are accessory treatments after reconstruction with LVA and lymphatic transfers

R EFERENCES 1. Smith JW, Conway H. Selection of appropriate surgical procedures in lymphedema. Introduction of the hinged pedicle. Plast Reconstr Surg Transplant Bull 30:10-31, 1962. 2. Thompson N. Buried dermal flap operation for chronic lymphedema of the extremities. Ten-year surgery of results in 79 cases. Plast Reconstr Surg 45:541-548, 1970. 3. Koshima I, Kawada S, Moriguchi T, et al. Ultrastructural observations of lymphatic vessels in lymphedema in human extremities. Plast Reconstr Surg 97:397-405; discussion 406-407, 1996. 4. Koshima I, Inagawa K, Urushibara K, et al. Supermicrosurgical lymphaticovenular anastomosis for the treatment of lymphedema in the upper extremities. J Reconstr Microsurg 16:437-442, 2000.

K23349_Neligan_35_Lymphatico Anastom_r3_dc_0473-0486.indd 485

5/27/2015 12:42:52 PM

486

Part V  Current Treatment of Lymphedema

5. Koshima I, Nanba Y, Tsutsui T, et al. Long-term follow-up after lymphaticovenular anastomosis for lymphedema in the leg. J Reconstr Microsurg 19:209-215, 2003. 6. Koshima I, Nanba Y, Tsutsui T, et al. Minimal invasive lymphaticovenular anastomosis under local anesthesia for leg lymphedema: is it effective for stage III and IV? Ann Plast Surg 53:261-266, 2004. 7. Degni M. New technique of lymphatic-venous anastomosis (buried type) for the treatment of lymphedema. Vasa 3:479-483, 1974. 8. O’Brien BM. Microlymphaticovenous anastomoses for obstructive lymphedema. Plast Reconstr Surg 60:197-211, 1977. 9. Degni M. New technique of lymphatic-venous anastomosis for the treatment of lymphedema. J Cardiovasc Surg 19:577-580, 1978. 10. Baumeister RG, Siuda S. Treatment of lymphedemas by microsurgical lymphatic grafting: what is proved? Plast Reconstr Surg 85:64-74; discussion 75-76, 1990. 11. Ogata F, Narushima M, Mihara M, et al. Intraoperative lymphography using indocyanine green dye for near-infrared fluorescence labeling in lymphedema. Ann Plast Surg 59:180-184, 2007. 12. Laine JB. Experimental lymphatico-venous anastomosis. Surg Forum 14:111-112, 1963. 13. O’Brien BM. Microlymphaticovenous anastomoses for obstructive lymphedema. Plast Reconstr Surg 60:197-211, 1977. 14. Yamada Y. Studies on lymphatic venous anastomosis in lymphedema. Nagoya J Med Sci 32:1-21, 1969. 15. Koshima I, Soeda S, Moriguchi T, et al. The use of arteriovenous anastomosis for replantation of the distal phalanx of the fingers. Plast Reconstr Surg 89:710-714, 1992. 16. Koshima I, Soeda S. Inferior epigastric artery skin flaps without rectus abdominis muscle. Br J Plast Surg 42:645-648, 1989.

K23349_Neligan_35_Lymphatico Anastom_r3_dc_0473-0486.indd 486

5/27/2015 12:42:52 PM

C hapter 36 Vascularized Lymph Node Transfer Corinne Becker

K ey P oints • Autologous vascularized lymph node transfer is the optimal treatment to reduce the swelling of lymphedema that is resistant to physical therapy. • Preoperative imaging studies to identify the leakage site are critical to the success of lymph node transfer. • To obtain good results, it is imperative that the pathology be well understood and evaluated so that the correct treatment strategy can be determined.

Vas

• MR lymphangiography helps identify the type of edema and is critical to deciding the best treatment option. • A combination of vascularized lymph node transfer and lymphaticovenular anastomosis is becoming more common and may yield superior results than either modality alone.

Lymphedema is a pathologic condition that results from a disturbance of the lymphatic system, with localized fluid retention and tissue swelling. Primary lymphedema is a congenital disorder caused by a malformation of lymph vessels and nodes. Chronic lymphedema is a progressive condition characterized by a degenerative and inflammatory deterioration of the dermis and subdermis, resulting in diffuse, irreversible fibrosis. Hypotrophy or hypoplasia of the lymphatic system occurs from a lack of lymphatic channel development or lymph nodes. In young adults with congenital lymphedema, swelling of the lower limb is unilateral in 70% of patients and bilateral in 30% of patients. Various forms of lymphedema affecting half of the body (right or left side) also exist. Major progress has been achieved in the radiologic diagnosis of patients with lymphedema. MR lymphangiography with T2-weighted imaging allows visualization of the anatomy of the lymphatic system with greater sensitivity than lymphoscintigraphy and potentially without the need for any injection,1,2 although the use of gadolinium with venous suppression probably yields more consistent images. The ideal treatment of the affected limb should restore both function and the cosmetic appearance. Physical therapy is the most commonly performed treatment for chronic lymphedema, and many consider it the only treatment for long-term management. Certainly, it helps to control to some extent the evolution of the disease, but it is not a curative therapy. 487

K23349_Neligan_36_Vascularized_r5_dc_0487-0502.indd 487

5/27/2015 12:41:21 PM

488

Part V  Current Treatment of Lymphedema

Interruption of regular therapy will invariably result in swelling of the affected segment and continued worsening of the condition. Lymph nodes express vascular endothelial growth factor C3, which promotes lymphangiogenesis. They also function as a sponge or “sump pump” for extracellular liquid, exchanging some lymphatic fluid with the venous circulation. Furthermore, they contain germinative cells.3,4 Lymphedema presents in various forms, and treatment is not the same for all patients. Physicians must distinguish between secondary or acquired lymphedema and congenital lymphedema. The former may occur because of filariasis, after various cancer treatments, and from trauma and some surgical procedures. The latter may appear at birth, during the first years of the life, after puberty, or even later. Autologous vascularized lymph node transfer (VLNT) is performed by some surgeons and is becoming increasingly popular. This technique is quite difficult to master and requires not only surgical and microsurgical skill, but also a good knowledge of the pathology involved. Although many of my cases are the result of combinations of treatment (surgery and radiation), they are labeled as acquired. Although they are acquired, they are also iatrogenic, which is not a label that is commonly used. Iatrogenic lymphedema can appear after lumpectomy or mastectomy combined with radiotherapy and lymphadenectomy. It appears in 20% to 60% of patients, sometimes months or years after those procedures, depending on the number of nodes removed and/or irradiated.5 The initial evaluation is done by lymphangioscintigraphy and recently by MR lymphangiography, which provides a real high-resolution map of the lymphatic system. If lymphatic drainage of the limb is completely absent, physical therapy will never resolve the situation. After excision of the scar tissue and placement of a free fatty flap containing some lymph nodes, VLNT is the best alternative. Excision of the scar tissue can be extremely difficult; it demands a great deal of experience and a detailed knowledge of the anatomy of the brachial plexus region (Fig. 36-1). The main indications for VLNT include recurrent infections, extreme fibrosis and even elephantiasis, pain, resistance to physical therapy confirmed by the absence of drainage

FIG. 36-1  Extreme fibrosis in the axillary region.

K23349_Neligan_36_Vascularized_r5_dc_0487-0502.indd 488

5/27/2015 12:41:22 PM

Chapter 36  Vascularized Lymph Node Transfer

489

on MRI, and in combination with breast reconstruction (Fig. 36-2). Although pain is not a major feature of lymphedema, it certainly occurs.6 Furthermore, patients often complain of discomfort and a feeling of extreme heaviness, which they sometimes equate with pain. The donor sites are the lateral inguinal region (eventually combined with a deep inferior epigastric perforator [DIEP] or superficial inferior epigastric artery [SIEA] flap) or lateral thoracic and cervical flaps, either submental of supraclavicular. The dissection is difficult and requires an indepth knowledge of the anatomy to minimize the risk of inducing lymphedema to the donor site. Iatrogenic lymphedema of the lower extremity can occur after tumor resection and lymphadenectomy in the pelvic area (hysterectomy, prostatectomy, and lymphadenectomy) and/or in the inguinal region, such as after lymphadenectomy for melanoma. It can also occur after nodal biopsies, saphenofemoral ligations, hernia repairs, or aesthetic and reconstructive procedures in the thigh (thigh lift, lipoaspiration, and transverse upper gracilis flaps). Identifying the problem is very important because it guides the surgeon on the optimal placement of the lymph nodes and which lymphatic pathways must be targeted during physical therapy. MR lymphangiography is the best investigation tool to evaluate the problem. The flap can be sometimes enlarged to reconstruct the breast defect and fibrotic area in the axillary or inguinal region. In our series, postoperative results showed improvement of between 40% for the upper extremity and 33% for the lower extremity in practically all patients, with complete resolution in a few patients after 2 years.7 The ultimate result depends on the quality of the skin, degree of underlying fibrosis, volume of the limb, number of infections (which destroyed the lymphatic vessels), age of the patient, and duration of the lymphedema. Patients with the best results no longer need compression sleeves or any type of ongoing therapy. In stage 2 or 3 lymphedema, improvement but not normalization was seen in 98% of patients.6 The infection rate, pain, and discomfort were significantly reduced. In rare cases of weakness or palsy associated with brachial plexopathy, stabilization of the palsy was observed, and after the volume was reduced, tendon transfers could subsequently be performed. Very localized lipo­ sculpture could be performed later after fat deposits were present.

FIG. 36-2  Absence of drainage on MRI. There is extensive lymphatic activity distal to the axilla, but no evidence of drainage in the axilla.

K23349_Neligan_36_Vascularized_r5_dc_0487-0502.indd 489

5/27/2015 12:41:22 PM

490

Part V  Current Treatment of Lymphedema

A successful outcome in stage 2 and 3 of the lower extremity was seen in about 50% of patients, but even patients with long-standing and extreme lymphedema had improvement.8 The worst results were observed in stage 3 and 4 bilateral lymphedema. Better results will be achieved the earlier the diagnosis and surgical intervention. Congenital lymphedema can present in various forms. Lymphoscintigraphy yields poor results and in our practice has been completely replaced by MR lymphangiography. However, this form of imaging is relatively new and is performed in only a few centers. The syndromes, hypoplastic or hyperplastic forms, and the level of the malformations are evaluated. Some individuals can pre­ sent simultaneously, with both hypoplastic and hyperplastic forms in different parts of the body. VLNT is indicated in patients with hypoplasia, because lymphangiogenic proteins are contained in the transplanted nodes, and they simultaneously act as sump pumps to transfer some of the lymphatic fluid into the venous circulation.8 Improvement was seen in most patients, even in those with advanced cases of elephantiasis. Mild cases, especially in children, can become normal. Combinations of VLNT with liposculpture can improve the aesthetic results by removing the fat deposits.8 In our center we treat both congenital and acquired forms of lymphedema, and we have reviewed our results. Two groups of patients were studied, one iatrogenic and the second congenital. During the past 20 years, more than 1500 cases have been reviewed. In upper extremity lymphedema, patients were classified based on the stage of the disease, duration of the lymphedema, number of infections, dash score, and associated problems, such as pain and palsy. In patients with lymphedema of the lower extremity, similar symptoms were examined. A multidisciplinary team from my center selected the candidates for surgery. Regardless of the patient’s previous treatments before referral for reconstructive surgery, disease progression for a minimum of 1 year was confirmed. Patients had preoperative evidence of substantial progression of lymphedema from clinical stage 1 to stage 2 or from stage 2 to stage 3 despite an adequate treatment program over at least 12 months or progressive lymph fluid accumulation as seen on MR lymphangiography (or by lymphoscintigraphy). For congenital cases, the hypoplastic forms were selected for VLNT. Sometimes combinations with other procedures were performed, mostly in patients with true elephantiasis. Elephantiasis can be caused by the hypoplastic or hyperplastic forms of the condition, and thus the diagnosis must be accurate. However, a combination of lymphatic reconstructive surgery with local excision was often necessary to avoid recurrent mycosis on the folds. Physical therapy was complementary to the surgical treatment. Compressive bandages were applied, and manual drainage was performed for the first 3 postoperative months and progressively reduced thereafter. Patients were evaluated by perimetry, volumetry, and quality of life, and preoperative and postoperative photographs were taken after 6 and 12 months and 2 and 5 years. Between 1992 and 2011, lymphoscintigraphy was performed preoperatively and 1 and 5 years postoperatively. However, the correlation with the clinical results was not always appropriate. Since 2011, MR lymphangiography has yielded superior information and excellent images of the lymphatic system.

K23349_Neligan_36_Vascularized_r5_dc_0487-0502.indd 490

5/27/2015 12:41:22 PM

491

Chapter 36  Vascularized Lymph Node Transfer

Surgical Technique for Upper Extremity Iatrogenic Lymphedema For upper extremity lymphedema, whether or not combined with modified DIEP for breast reconstruction, the surgical approach to the axilla was performed through the previous scar. The scar tissue was dissected and adhesions were released as far as the anterior border of the latissimus dorsi. Dissection then continued until the thoracodorsal vessels and subscapular system were identified. This dissection can be very difficult, particularly after radiotherapy, because all the vessels and brachial plexus can be involved in the scar. Injury to these structures is a significant hazard. For this reason, surgical experience and anatomic knowledge are very important. After all the scar tissue was removed, the flap was placed in the apex of the axilla around these vessels. Microvascular anastomosis was made to available vessels, quite commonly the thoracodorsals.

Lateral Inguinal Flap The lateral inguinal flap is supplied by the superficial circumflex iliac vessels, and anastomosis with the deep inferior epigastric vessels was seen in 80% of patients. An oblique incision was made between the iliac crest and the pubis above the inguinal crease (Fig. 36-3, A). The superficial circumflex iliac vessels were identified. The flap was raised on these vessels and included four to five nodes (Fig. 36-3, B). These nodes were dissected, freed, and elevated from lateral to medial

A

Inguinal ligament Branches of deep circumflex iliac vessels

B Flap margin Iliac crest

Superior inferior epigastric vessels

Incision above inguinal crease

Inferior epigastric vessels (deep) Pubis Femoral vessels

Lymph nodes to be transferred (4-5 maximum) Superficial circumflex iliac vessels

FIG. 36-3  A, An oblique incision is made between the iliac crest and pubis above the inguinal crease. The superficial circumflex iliac vessels are identified. B, The flap is raised on these vessels and includes four to five nodes. These nodes are dissected, freed, and elevated from lateral to medial at the level of the iliac crest. The flap can be extended medially if more bulk is required. It is important to note that the inguinal fold must be the lower limit of the flap and not deeper into the fascia. Pulling is very dangerous.

K23349_Neligan_36_Vascularized_r5_dc_0487-0502.indd 491

5/27/2015 12:41:22 PM

492

Part V  Current Treatment of Lymphedema

at the level of the abdominal wall fascia extending medially if more bulk was required. The inguinal fold must be the lower limit of the flap. If excessive dissection is performed in the thigh, the risk of lymphedema at the donor site is significant. The dissection began by visualization of the superficial circumflex iliac vein. In that region, some lymph nodes are supplied by the circumflex iliac vessels, which mainly drain lymph from the abdominal wall. For this reason the harvesting of this flap does not impair the lymphatic drainage of the lower limb. This flap can be transferred with a DIEP or SIEA, or it can be added as a free flap on a branch of another flap, such as the thoracodorsal artery perforator, superior artery gluteal perforator, or inferior gluteal artery perforator, or to a separate donor vessel in the axilla9 (Fig. 36-4). The circumflex iliac vessels must be dissected as far as possible to achieve maximal length and caliber. The DIEP can incorporate this flap, but it is important to confirm that the lymph node flap is well vascularized. In the axillary region, debridement of the scar tissue and individualization of the branches of the thoracodorsal vessels were performed first. If the DIEP anastomosis is done to the mammary vessels, the VLNT must be dissected on the side opposite the DIEP pedicle. In this situation, it should not be assumed that the vascularity of the flap is carried by the DIEP. In most cases, a separate pedicle should be dissected for the lateral inguinal flap. In this case the flap will be anastomosed as the second anastomosis in the axilla (Fig. 36-5). A second anastomosis was preferred to improve the vascularization of the transplanted nodes. Drains were placed in the defect, which was closed in two layers. The postoperative care of the

FIG. 36-4  VLNT extended in the SIEA.

FIG. 36-5  MR lymphangiography showing the flap inserted in the axillary region.

K23349_Neligan_36_Vascularized_r5_dc_0487-0502.indd 492

5/27/2015 12:41:22 PM

Chapter 36  Vascularized Lymph Node Transfer

493

inguinal region was important. The drains were removed after the second day, and a compression garment such as Spanx was recommended to prevent lymphocele. Perioperative antibiotic therapy was sufficient to prevent infection.

Surgical Technique for Lower Extremity Iatrogenic Lymphedema Depending on the cause and extent of the lymphedema, the flap may be placed in different locations. If the flap is placed in the groin, dissection of scar tissue around the femoral and inguinal vessels in heavily irradiated tissue is difficult, and great care must be taken to avoid injury to these vessels. Sometimes if the patient complains of pain, neurolysis of the crural and femoral nerves is needed. Recipient vessels can then be identified and used for the microanastomosis. In very extensive edema, a second flap will usually be inserted around the knee region, but it can also be inserted more distally in the calf or near the ankle, depending on the growth of the lymphatic vessels as assessed by MRI some months after the first inguinal flap. The donor flaps were the lateral thoracic flap and supraclavicular flap.10

Lateral Thoracic Flap The incision was performed behind the lateral border of the pectoralis major muscle. The surgeon must stay away as far as possible from the apex of the axilla to avoid injury to the axillary nodes draining the arm. The branches supplying the three or four nodes were identified, and these generally were from the serratus branch of the subscapular system or thoracodorsal artery. Exposure of the axillary vein was avoided during surgery. The dissection was performed, and the apical nodes were left undisturbed. A thorough knowledge of the anatomy is extremely important, and I do not recommend dissection proximally on the thoracodorsal vessels (Fig. 36-6). No traction should be exerted during the dissection that

FIG. 36-6  Lateral thoracic flap. Note branches from the thoracodorsal vessel, which supplies the lateral thoracic nodes.

K23349_Neligan_36_Vascularized_r5_dc_0487-0502.indd 493

5/27/2015 12:41:22 PM

494

Part V  Current Treatment of Lymphedema

could pull the contents of the apex of the axilla into the direct field so that no damage occurs to the apical lymph nodes. It is very easy to dissect too deep in the upper part of the axilla. The closure of the axillary region was performed in two layers over a drain that was removed after 2 days.

Cervical Flap The incision was performed along the upper part of the clavicle. The nodes, which are supplied by the transverse cervical artery, were identified behind the sternocleidomastoid muscle lateral to the internal jugular vein. The external jugular vein can be preserved during the dissection, although it is not an issue if it is sacrificed. The vascular pedicle is on the deep surface of the flap, and it is easiest to identify the transverse cervical vessels medially. Knowledge of the anatomy of the neck is important to avoid damage to vital structures, such as the thoracic duct on the left side and the right lymph duct on the right side (Fig. 36-7).

A

B

Sternocleidomastoid muscle

External jugular vein Internal jugular vein Omohyoid muscle Transverse cervical vessels Right lymphatic duct Incision

Flap margin Supraclavicular nodes to be transferred (6 minimum)

FIG. 36-7  The transverse cervical artery supplies nodes just at the lateral border of the sternocleidomastoid muscle, above the clavicle.

K23349_Neligan_36_Vascularized_r5_dc_0487-0502.indd 494

5/27/2015 12:41:23 PM

Chapter 36  Vascularized Lymph Node Transfer

495

Surgical Technique for Congenital Lower Limb Lymphedema The recipient sites were chosen based on MR lymphangiography. If the lymphedema involves the whole leg, two flaps will be necessary. One flap will be inserted in the inguinal region, usually on the circumflex iliac vessels, and the second flap will be inserted more distally in the lower leg in the calf or near the ankle, depending on the images of the MR lymphangiography. Once again, the donor sites were the lateral thoracic or supraclavicular flap. Drains were placed at both sites for 2 days. Intraoperative antibiotic therapy was used. The patient’s stay at the hospital was generally 24 to 48 hours.8

Results For iatrogenic stage 1 and 2 lymphedema after mastectomy and radiotherapy, 98% of the patients evaluated at 1 year showed clinical improvement, with an average reduction in limb volume of 50%11 (Fig. 36-8). All patients had clinical improvement with fewer episodes of sepsis. However,

A

C

B

D

FIG. 36-8  A, This patient had a 15-year history of stage 2 lymphedema of the left arm after mastectomy, lymphadenectomy, and radiotherapy. B, Postoperative result. C and D, Preoperative and postoperative evaluation with lymphoscintigraphy.

K23349_Neligan_36_Vascularized_r5_dc_0487-0502.indd 495

5/27/2015 12:41:23 PM

496

Part V  Current Treatment of Lymphedema

the correlation with lymphoscintigraphy was poor (Fig. 36-9). The MR lymphangiography showed new lymphatic vessels in 50% of patients, and all patients had transplanted nodes and a reduction of stasis.11 For patients with stage 3 and 4 disease, significant improvement was shown, with a reduction of approximately 60% in limb volume and a dramatically reduced rate of infections. However, in these patients complete normalization was never observed and physical therapy must continue. MR lymphangiography showed less dermal backflow and new lymphatic vessels. Interestingly, the honeycombs of fat seen before surgery disappeared, but there was still greater fat deposition than in the contralateral arm. Therefore very localized and selective liposuction of the lateral and posterior aspects of the arm can achieve better aesthetic results. Care was taken to avoid suction of the medial aspect of the arm. Postoperative compression for these minimal liposculpture procedures was only needed for 4 weeks, unlike the requirement of continuous compression (around the clock, 7 days a week) as in the Brorson technique (Fig. 36-10).

FIG. 36-9  Clinical results after 5 years. Lymphoscintigraphy showed new pathways and nodes. Ten years later, MR lymphangiography showed complete normalization.

A

B

FIG. 36-10  A, This patient had a 30-year history of right arm elephantiasis, with frequent infections. She was treated with two free flaps, one in the axillary region and the second at the elbow region. B, Results are shown after 4 years.

K23349_Neligan_36_Vascularized_r5_dc_0487-0502.indd 496

5/27/2015 12:41:23 PM

Chapter 36  Vascularized Lymph Node Transfer

497

For patients with stage 2 secondary lymphedema of the lower extremity, the results showed clinical improvement in 50% of patients, with no further episodes of sepsis and improved quality of life.12 Twenty percent of patients reverted to normal (Fig. 36-11). In patients with stage 3 and 4 disease of the lower extremity, improvement was seen in 30% of cases.12 The number of infections decreased significantly in all patients (Fig. 36-12).

A

B

FIG. 36-11  A, This patient had an 8-year history of lymphedema of the left lower extremity. B, Results are shown 2 years after VLNT to the deep inguinal region.

A

B

FIG. 36-12  A, This patient had a 25-year history of lymphedema with chronic infections, treated with two VLNTs. The first VLNT was in the inguinal region, and the second was in the region of the knee 6 months later. B, Results are shown after 2 years.

K23349_Neligan_36_Vascularized_r5_dc_0487-0502.indd 497

5/27/2015 12:41:24 PM

498

Part V  Current Treatment of Lymphedema

A

B

FIG. 36-13  A, This patient had lymphedema of the right lower extremity 7 years after treatment for melanoma. B, Postoperative result.

In patients with bilateral involvement after pelvic resection combined with resection of the deep iliac and inguinal nodes and postoperative radiotherapy, the remaining pathways were sometimes nonexistent. The flaps were inserted in both groins and subsequently at knee level. In these patients the results showed a reduction in the number of infections and a volume reduction of 20% after 1 year.12 MR lymphangiography gave valuable information on optimal node placement (Fig. 36-13).

Results One Year After Two Vascularized Lymph Node Transfers If lymphedema occurs after saphenous vein ligation, lymphadenectomy, or thigh lift, the flap should be inserted exactly at the location of the scar. The results are always better if the fibrosis is minimal and the lymphedema has not been present for many years. Additional liposculpture can be performed later after the lymphedema has become transformed into lipedema again, confining the liposuction to the lateral and posterior segments and avoiding the medial thigh where the lymphatic collectors are situated. Minor complications occurred in more than one third of the patients analyzed. These complications included seroma, temporary pain after surgery, and hematoma. Lymphedema of the donor site was very rare (1% incidence).3 Although this complication was rare, it supports the concept

K23349_Neligan_36_Vascularized_r5_dc_0487-0502.indd 498

5/27/2015 12:41:24 PM

Chapter 36  Vascularized Lymph Node Transfer

A

499

B

FIG. 36-14  A, This 19-year-old girl presented with hypoplasia of the inguinal region on the left side and hypoplasia of both legs distally. B, Postoperative results.

that careful dissection of the flap must be carried out, avoiding damage to the more medial part of the lymphatic system. The development of surgical strategies to reduce these risks is important. For congenital cases, evaluation of the results is relatively difficult to standardize because of the presence of bilateral forms, the growth of children, and weight gain in adults: • Group 1 (distal lymphedema): All patients in this group showed a reduction of the circumference of the treated limb compared with the contralateral side, with normalization in 46% of patients. Infections disappeared in 88% of patients. MR lymphangiography showed the new lymphatic pathways. • Group 2 (generalized limb lymphedema): The measurements were done in the same way. Improvement was seen in 98%, of patients, but only 20% had normalization. In extensive and more long-standing lymphedema, a second flap placed in the knee region at the confluence of the deep and superficial pathways seemed to improve the results. The rate of infections decreased dramatically. However, 53% of patients had recurring erysipelas. The result after placement of one flap in the left inguinal region and around the knee was that the left leg improved compared with right leg. Physical therapy was no longer performed on this patient, but perhaps it would have improved the result (Fig. 36-14).

K23349_Neligan_36_Vascularized_r5_dc_0487-0502.indd 499

5/27/2015 12:41:24 PM

500

Part V  Current Treatment of Lymphedema

A

B

FIG. 36-15  A, This 6-month-old baby had lymphedema of the right lower extremity. B, Results 12 months after VLNT combined with miniresection of the mass on the foot.

An infant with lymphedema of the right lower extremity is shown in Fig. 36-15.

Discussion Autologous VLNT is the best treatment to improve swelling that is resistant to physical therapy, even in patients with extensive lymphedema. The transplanted lymph node can release vascular endothelial growth factor C that stimulates lymphangiogenesis. The transplanted nodes can also act as a sponge or sump pump to absorb lymphatic fluid in the edematous limb. The transplanted lymph node can help improve the immune function of the edematous limb and thereby reduce the risk of cellulitis. In iatrogenic cases, the release of scar tissue and importation of good vascularized fat must also be considered. A more objective evaluation of the efficacy of the procedure can be appreciated by a serial volume comparison of the affected limb with the healthy limb. In a series of lower limb iatrogenic lymphedema, moderate (more than 30% excess volume reduction) to good results (more than 75% excess volume reduction) were seen. Better results were also observed in patients with less severe forms of lymphedema (excess volume less than 50%). The site of lymphatic blockage is less predictable in patients with congenital lymphedema compared with iatrogenic lymphedema (for example, the axilla after axillary dissection and the groin after pelvic or groin dissection). Therefore preoperative imaging studies to identify the site of blockage are crucial to the success of the lymph node transfer. The new technology of MR lymphangiography helps identify the type of edema: hypoplasia of the nodes and vessels, the level of hypoplasia, and other different forms.

K23349_Neligan_36_Vascularized_r5_dc_0487-0502.indd 500

5/27/2015 12:41:24 PM

Chapter 36  Vascularized Lymph Node Transfer

501

If the pathology is well understood and evaluated, the treatment strategy can be determined, and this is imperative to obtain good results. The multidisciplinary approach (for example, physical therapist, radiologist, and histologist) allows a better understanding of the disease process and ensures that the right treatment will be chosen. The use of VLNT in hypoplastic lymphedema is the technique of choice. It is a logical approach for the reconstruction of a damaged lymph transport system. Derhy et al1 described the use of MR lymphangiography to visualize very precisely the lymphatic pathways without any injection. This examination is the key to deciding the best treatment option. In patients with lesions of the thoracic duct or hyperplasia, lymphaticovenous anastomosis seems to be indicated, but the detection of good quality lymphatic vessels is important. Indocyanine green lymphangiography is useful in this regard.13 In the late stages of elephantiasis, the channels are obstructed, and local resections are needed to remove the folds and avoid mycosis. For a surgeon well trained in microsurgery, the failure rate does not exceed 2%. The morbidity from VLNT includes donor site seroma and hematoma. Donor site lymphedema is the worst possible complication, but fortunately it is very rare. If the lymphedema is not too advanced, complete or near-complete recovery is possible. The success of such operations depends on good collaboration between biologists, radiologists, reconstructive surgeons, and physical therapists. In the future there will be combinations of different techniques: local excision of the folds if necessary, VLNT, and later localized liposculpture. Lymphaticovenular anastomosis is reserved for hyperplastic cases but can be used in the ankle regions to finish the aesthetic result in selected patients.14 The combination of VLNT and lymphaticovenular anastomosis is becoming more commonplace and may yield superior results than either modality alone. Of course, functioning lymphatics must be present to ensure the success of lymphaticovenous anastomosis. One area of controversy is the placement of the VLNT. As previously mentioned, my approach is to place the nodes at the site of nodal excision in iatrogenic cases; care is taken to excise the existing scar tissue. In congenital cases it is less clear-cut, and my approach has been to place the nodes at the site of blockage as identified on MRI. To the contrary, Yamamoto et al15 place the nodes in the most distal and thus the most dependent part of the limb, because the lymph nodes act as sponges or sump pumps and should therefore be placed in the area of maximal edema.

C linical P earls • Preoperative MR lymphangiography is a vital part of the patient workup. • When harvesting lateral inguinal nodes, it is important not to dissect below the groin crease. • Patients may need to be supplemented with a second VLNT to further improve their condition.

K23349_Neligan_36_Vascularized_r5_dc_0487-0502.indd 501

5/27/2015 12:41:24 PM

502

Part V  Current Treatment of Lymphedema

R EFERENCES 1. Derhy S, Mouhadi E, Ruiz S, et al. Non-contrast 3D MR lymphography of retroperitoneal lymphatic aneurysmal dilatation: a continuous spectrum of change from normal variants to cystic lymphangioma. Insights Imaging 4:753-758, 2013. 2. Arrivé L, Azizi L, Lewin M. MR lymphography of abdominal and retroperitoneal lymphatic vessels. Am J Roentgenol 189:1051-1058, 2007. 3. Viitanen TP, Mäki MT, Seppänen MP, et al. Donor-site lymphatic function after microvascular lymph node transfer. Plast Reconstr Surg 130:1246-1253, 2012. 4. Lähteenvuo M, Honkonen K, Tervala T, et al. Growth factor therapy and autologous lymph node transfer in lymphedema. Circulation 123:613-620, 2011. 5. Suami H, Chang DW. Overview of surgical treatments for breast cancer-related lymphedema. Plast Reconstr Surg 126:1853-1863, 2010. 6. Becker C, Pham DN, Assouad J, et al. Postmastectomy neuropathic pain: results of microsurgical lymph nodes transplantation. Breast 17:472-476, 2008. 7. Becker C, Arrivé L. Congenital lymphedema clinical results compared with lympho-MRI (in press). 8. Becker C, Arrivé L, Saaristo A, et al. Surgical treatment of congenital lymphedema. Clin Plast Surg 39:377-384, 2012. 9. Assouad J, Becker C, Hidden G, et al. The cutaneo-lymph node flap of the superficial circumflex artery. Surgical and radiologic anatomy. Surg Radiol Anat 24:87-90, 2002. 10. Becker C, Hidden G. [Transfer of free lymphatic flaps. Microsurgery and anatomical study] J Mal Vasc 13:119-122, 1988. 11. Becker C, Assouad J, Riquet M, et al. Postmastectomy lymphedema: long-term results following microsurgical lymph node transplantation. Ann Surg 243:313-315, 2006. 12. Batista BN, Becker C, Germain M. Vascularized lymph node transfer for patients with secondary inferior limb lymphedema. Plast Reconstr Surg 132(4S-1):32-33, 2013. 13. Brorson H, Höijer P. Standardised measurements used to order compression garments can be used to calculate arm volumes to evaluate lymphoedema treatment. J Plast Surg Hand Surg 46:410-415, 2012. 14. Koshima I, Nanba Y, Tsutsui T, et al. Minimal invasive lymphaticovenular anastomosis under local anesthesia for leg lymphedema: is it effective for stage III and IV? Ann Plast Surg 53:261-266, 2004. 15. Yamamoto T, Matsuda N, Doi K, et al. The earliest finding of indocyanine green lymphography in asymptomatic limbs of lower extremity lymphedema patients secondary to cancer treatment: the modified dermal backflow stage and concept of subclinical lymphedema. Plast Reconstr Surg 128:314e-321e, 2011.

K23349_Neligan_36_Vascularized_r5_dc_0487-0502.indd 502

5/27/2015 12:41:24 PM

C hapter 37 Reverse Lymphatic Mapping Joseph H. Dayan, Mark L. Smith

K ey P oints • The risk of donor-site lymphedema is relevant to the harvest of groin, axillary, and supraclavicular lymph nodes. • Reverse lymphatic mapping enables the surgeon to identify lymph nodes draining the extremity so that they can be avoided during lymph node transfer. • Anatomic landmarks do not always correlate with physiologic lymphatic drainage patterns, which can be identified with reverse lymphatic mapping. • Reverse lymphatic mapping provides physiologic information that can alter the surgical plan and improve the safety of lymph node transfer.

Rev

Vascularized lymph node transfer (VLNT) has become increasingly popular for the treatment of lymphedema, because it is conceptually appealing and uses techniques familiar to every microsurgeon. Although VLNT has been effective in the treatment of lymphedema, the potential for creating lymphedema at the donor site is the primary concern of both the surgeon and patient. Reports of donor-site lymphedema have surfaced in the literature, even when surgery was performed by very experienced microsurgeons with the use of traditionally accepted anatomic landmarks.1-5 This risk is a real one, and at the same time, it would be premature to dismiss VLNT entirely, because it has yielded significant improvement in numerous patients who had previously coped with a disabling and unrelenting disease.6-8 The question now becomes, how can we perform this technique while minimizing the potential risk of donor-site lymphedema? We developed reverse lymphatic mapping for VLNT to specifically address the risk of donor-site lymphedema. We have used reverse lymphatic mapping to harvest lymph nodes from the inguinal, axillary, and supraclavicular regions to avoid harvesting lymph nodes that drain the adjacent limb.

503

K23349_Neligan_37_Reverse Lymph_r4_dc_0503-0512.indd 503

5/27/2015 12:44:20 PM

504

Part V  Current Treatment of Lymphedema

Concept Reverse mapping was first conceived by Thompson et al9 and Klimberg10 to reduce the risk of iatrogenic lymphedema after sentinel lymph node biopsy or axillary lymphadenectomy for breast cancer treatment. This technique, described as axillary reverse mapping, involves the injection of blue dye in the upper extremity and technetium in the breast. Avoiding blue lymph nodes that drain the upper limb during sentinel lymph node biopsy or axillary lymphadenectomy (unless they share drainage with the breast) potentially reduces the risk of developing lymphedema. We11 have applied this concept and modified it for VLNT to minimize the risk of donor-site lymphedema. Technetium is injected into the hand or foot, depending on where the donor site of the lymph node flap is located. A gamma probe is used intraoperatively to locate the extremitydraining lymph nodes, which are then avoided. Indocyanine green is also injected into the trunk, and with near-infrared imaging, the target lymph nodes draining the trunk can be identified and included in the flap. This approach provides valuable physiologic information that can alter flap harvest, as opposed to solely using anatomic landmarks as a guide. We have observed several aberrant lymphatic drainage patterns with reverse lymphatic mapping. By using reverse lymphatic mapping, we have identified and avoided lymph nodes draining the leg that typically would have been harvested if anatomic landmarks alone were used, thus potentially reducing the risk of iatrogenic lymphedema. One limitation that we encountered in our early experience with blue dye injection was that the lymph nodes could only be seen when directly exposed in the operative field. At this point, the delicate lymphatic network and vascular supply may be jeopardized, and harvesting compromised lymph nodes may lead to a failed lymph node transfer. The use of indocyanine green lymphangiography with a near-infrared detector allowed us to see the lymph nodes before they were directly exposed in the field, which enabled the preservation of the lymph nodes in the flap with maximum viability.

Reverse Lymphatic Mapping in Vascularized Groin Lymph Node Transfer Anatomy The superficial inguinal lymph nodes are typically harvested based on the superficial circumflex iliac artery (SCIA). This artery commonly arises from the common femoral artery in the femoral triangle, although it may sometimes have an anomalous takeoff from the deep circumflex iliac artery (DCIA). The venous drainage of this flap can be through the venae comitantes or an alternative superficial vein.

K23349_Neligan_37_Reverse Lymph_r4_dc_0503-0512.indd 504

5/27/2015 12:44:20 PM

Chapter 37  Reverse Lymphatic Mapping

A

ASIS

505

B

SCIV SCIA Inguinal ligament

SIEV 1

Lymph nodes for transfer

∕3 distance from PT to ASIS

3 cm

SFA

Sartorius muscle

PT

GSV

SFV

FIG. 37-1  A, Anatomic landmarks to identify superficial inguinal lymph nodes for harvest. (ASIS, Anterior superior iliac spine; GSV, great saphenous vein; PT, pubic tubercle; SCIA, superficial circumflex iliac artery; SCIV, superficial circumflex iliac vein; SIEV, superficial inferior epigastric vein; SFA, superficial femoral artery; SFV, superficial femoral vein.) B, Intraoperative markings for vascularized groin lymph node transfer.

Based on our previous anatomic study,12 the SCIA-based lymph nodes are concentrated at the junction of the superficial circumflex iliac and superficial inferior epigastric veins. Anatomically, this corresponds to the region between the inguinal ligament and groin crease, which is approximately one third the distance from the pubic tubercle toward the anterior superior iliac spine (Fig. 37-1). The superficial lymph nodes lie just deep to Scarpa’s fascia. Although these nodes predominantly drain the lower abdomen, on occasion there can be SCIA-based lymph nodes that significantly contribute to lower extremity drainage, and these can be identified with reverse lymphatic mapping. Lymph nodes below the groin crease consistently drain the lower limb and should be avoided.

Surgical Technique Approximately 1 hour before surgery, the patient is injected with 0.2 ml of filtered technetium sulfur colloid (0.2 mCi/0.2 ml) in the first and second web spaces of the foot on the same side as the planned donor site. After the patient is prepared and draped, 0.1 ml of indocyanine green is injected intradermally across the lower abdomen in four areas halfway between the region of the umbilicus and inguinal ligament. The area is massaged to facilitate flow of the dye into the lymphatics and inguinal lymph node basin (Fig. 37-2).

K23349_Neligan_37_Reverse Lymph_r4_dc_0503-0512.indd 505

5/27/2015 12:44:21 PM

506

Part V  Current Treatment of Lymphedema

©2014 Mount Sinai Health System

Indocyanine green Groin crease Target nodes Nodes to avoid

FIG. 37-2  Injection sites for reverse lymphatic mapping in the lower limb.

Technetium

At this point, the inguinal crease is marked, as well as the course of the inguinal ligament between the pubic tubercle and anterior superior iliac spine. A handheld Doppler is used to confirm the course of the SCIA and superficial inferior epigastric artery. The gamma probe is now used to locate the sentinel lymph node draining the lower limb, which typically lies along the femoral vessels slightly below the inguinal crease. There will always be a signal in the region of the flap before dissection, because the probe will pick up the technetium in the deeper iliac lymph nodes. Thus this should not be a cause for concern. Near-infrared imaging is now used to visualize the lower abdominal lymphatic vessels and the corresponding lymph nodes targeted for harvest. Because the lymph nodes are deeper, these nodes will appear as a faint blush but will become obvious once the flap elevation begins. Most commonly, we harvest a lymph node flap without a skin island from the groin for use in the treatment of breast cancer–related lymphedema. The skin is incised over the SCIA, and subcutaneous flaps are elevated superficial to Scarpa’s fascia, which exposes the superficial surface of the lymph node flap. The near-infrared imaging is used once again to confirm the location of the target nodes. The flap is then elevated from lateral to medial, typically superficial to the sartorius muscle fascia. There can be variations in the origin and course of the SCIA. Therefore preoperative imaging may be beneficial. During lateral-to-medial flap elevation, the gamma probe is placed on the undersurface of the flap and aimed toward the ceiling to avoid signal artifact from the lower limb or deeper iliac nodes. This is a critical step in confirming the lack of signal in the lymph nodes based on the SCIA. If there is a high signal present, the decision is typically made to abort the flap and either to stop or switch to an alternate donor site. We secure consent from

K23349_Neligan_37_Reverse Lymph_r4_dc_0503-0512.indd 506

5/27/2015 12:44:21 PM

Chapter 37  Reverse Lymphatic Mapping

507

FIG. 37-3  Vascularized groin lymph node flap inset into the axilla with anastomosis to the thoracodorsal vessels.

all patients for an alternative donor site if such a scenario occurs. On two occasions to date we have found significant technetium-99 uptake from the lower extremity by an SCIA-based lymph node, which resulted in a modification of the procedure or the use of an alternate site. The patient should be informed before surgery regarding the potential need for an alternate approach if this situation arises. After the pedicle is identified, the next decision is to determine the medial border of the flap. This is an important point in the case, because there are medial lymph nodes in the same dissection plane that do significantly contribute to lower extremity drainage and are typically medial to the superficial inferior epigastric vein. However, as mentioned previously, static landmarks can be misleading. Thus the gamma probe is used again to interrogate the more medial lymph nodes. The surgeon takes the time necessary to make this decision; the goal is to capture as many viable nodes as possible without sacrificing drainage to the lower limb. If there is any doubt, the more conservative route is always taken and the lymph node is spared. An unsuccessful lymph node transfer is clearly preferable to iatrogenic lymphedema. Finally, the medial border of the flap is then defined, and the flap dissection is completed. After flap harvest, a 10-second count of the sentinel lymph nodes of the lower extremity and lymph node flap is separately recorded for documentation and further study. Near-infrared imaging can be used to assess the content of the lymph nodes in the flap if desired. The flap is then transferred, and intravenous indocyanine green is administered after anastomosis to confirm good perfusion of the flap (Fig. 37-3).

Reverse Lymphatic Mapping in Axillary Lymph Node Transfer Anatomy The lymph nodes targeted for transfer from the axillary donor site can be based on the thoracodorsal vessels and/or the lateral thoracic vessels. There are anatomic variations in the origin of the lateral thoracic artery, including the possibility of an absent lateral thoracic artery. These

K23349_Neligan_37_Reverse Lymph_r4_dc_0503-0512.indd 507

5/27/2015 12:44:21 PM

508

Part V  Current Treatment of Lymphedema

Technetium Nodes to avoid Target nodes Indocyanine green

©2014 Mount Sinai Health System

FIG. 37-4  Injection sites for reverse lymphatic mapping in the upper limb.

variations clearly have a role in determining the vascular pedicle or pedicles of the harvested flap and can sometimes require turbocharging or supercharging techniques if the lymph node flap has a dual blood supply completely dependent on both the lateral thoracic and thoracodorsal vessels. Preoperative MR angiography or other imaging, such as ultrasound, can identify the side with the most inferiorly located lymph nodes. A crop of lymph nodes significantly inferior to the axillary fold is most favorable, because this is farthest from the lymph nodes draining the upper extremity, which are situated high in the axilla along the axillary vein.13

Surgical Technique One hour before surgery, 0.2 ml aliquots of filtered technetium-99 sulfur colloid are injected into the first and second web spaces of the hand on the side of the lymph node harvest. After the patient is under anesthesia, a series of five 0.1 ml intradermal injections of indocyanine green is administered from the anterior axillary line to the back over the latissimus muscle. These are oriented transversely approximately 10 to 15 cm below the axillary fold. The area is massaged toward the axillary lymph nodes draining the trunk (Fig. 37-4). The patient is prepped and draped, allowing time for the indocyanine green to transit to the target nodes. The gamma probe is then used to localize the sentinel lymph node draining the upper extremity. This is typically high in the axilla, immediately underneath the superior edge of the pectoralis major muscle. Near-infrared imaging is then used to visualize the lymphatic vessels draining the trunk as they coalesce toward the target lymph nodes. This imaging is helpful to determine the ideal location of the incision, which should lie a safe distance from the sentinel lymph nodes of the upper extremity. Most of our flap procedures do not include a skin paddle unless skin resurfacing is required. In these cases, the skin paddle is based on the thoracodorsal artery perforator, which is typically inferior to the perforators supplying the lymph nodes and significantly increases the complexity of the case. For purposes of clarity, the following describes the use of reverse lymphatic mapping for a lymph node–only flap without a skin paddle.

K23349_Neligan_37_Reverse Lymph_r4_dc_0503-0512.indd 508

5/27/2015 12:44:21 PM

Chapter 37  Reverse Lymphatic Mapping

509

The axillary crease, pectoralis major muscle, and anterior border of the latissimus muscle are marked (Fig. 37-5). A transverse incision is made within these muscle boundaries over the area of the target nodes and should be inferior to the axillary crease. A vertical incision can also be used and may provide more direct visualization but often results in a hypertrophic scar. Subcutaneous flaps are elevated, and near-infrared imaging is used to localize the target lymph nodes. The gamma probe is used again to confirm the location of the sentinel nodes of the upper extremity, and the superior extent of the flap dissection is marked. The area is palpated to confirm the presence of lymph nodes and to evaluate how anterior or posterior the target nodes are situated. Nodes situated along the lateral chest wall are often supplied by the lateral thoracic artery, whereas nodes on the deep surface of the latissimus muscle are supplied by the thoracodorsal artery. The decision to use one or both of these vessels completely depends on the patient’s anatomy. After the distal pedicle is identified, the flap is elevated superiorly, constantly checking for technetium uptake with the gamma probe. The most critical decision is determining the superior border of the flap. The gamma probe is vital to make this decision, which is often a careful and tedious process because of the lack of consistency in the axilla and the loose perinodal fat that can easily change its orientation. After the superior extent of the flap is developed and dissected, the pedicle is teased from the thoracodorsal nerve, which is preserved. If the nerve is entangled between the perforators supplying the lymph nodes, the nerve is divided and then repaired under the microscope. Great care is taken to preserve the delicate perforators supplying the lymph nodes. The flap is then harvested, and a 10-second count of the flap and sentinel node of the upper limb is taken for documentation (Fig. 37-6).

FIG. 37-5  Intraoperative markings for vascularized axillary lymph node transfer based on the lateral thoracic and/or thoracodorsal vessels. The transverse incision is made well below the axillary crease.

FIG. 37-6  Axillary lymph node flap based on the thoracodorsal artery and vein and lateral thoracic vein.

K23349_Neligan_37_Reverse Lymph_r4_dc_0503-0512.indd 509

5/27/2015 12:44:22 PM

510

Part V  Current Treatment of Lymphedema

Reverse Lymphatic Mapping in the Supraclavicular Region There is a risk of upper extremity lymphedema after flap harvest of the supraclavicular lymph nodes. Although we have not observed high levels of technetium uptake in the supraclavicular nodes after technetium injection into the hand, we have come across some interesting findings that have altered our surgical technique after indocyanine green injection into the upper limb. When we harvest a supraclavicular flap, 0.2 ml of indocyanine green is injected into the medial upper arm, followed by massage. After incision and exposure of the supraclavicular lymph nodes, near-infrared imaging is used to look for lymph nodes with significant uptake of indocyanine green and to identify the right lymphatic or thoracic duct, depending on the side harvested. Major lymphatic ducts can resemble veins and may be misleading but are readily identified with indocyanine green lymphangiography. We saw one patient in whom the medialmost nodes surrounding the transverse cervical artery pedicle had significant drainage from the upper limb. A more lateral section of the lymph node flap was harvested, and the distal transverse cervical artery was anastomosed to the recipient artery.

Conclusion Although the benefits of lymph node transfer have been demonstrated, the risk of donor-site lymphedema is the primary reason that this technique has not yet been widely adopted. Reverse lymphatic mapping is a positive step in the evolution of this technique and provides a safer means for flap harvest. Reverse lymphatic mapping provides a physiologically based approach to harvesting a physiologic organ. The “augmented reality” gained by the use of technetium and nearinfrared imaging enables the surgeon to negotiate the potentially complex minefield of critical lymph nodes and to cherry pick nonessential nodes with greater confidence. As we increase our understanding of the anatomy and physiology of the lymphatic system, the safety and reliability of these procedures will undoubtedly continue to improve.

C linical P earls • MR angiographic imaging is helpful to assess donor-site and recipient-site anatomy. Information regarding the number and location of lymph nodes and their relationship to nutrient vessels can be visualized before surgery and help with surgical planning. • Ideally, the donor site is chosen from the contralateral side of the body to preserve lymph node basins that are adjacent to the affected extremity. However, anatomic consideration may supersede this general rule. • The inguinal donor site is prone to seroma formation. In addition to drain placement, quilting sutures can help facilitate tissue adherence and obliteration of the donor cavity.

K23349_Neligan_37_Reverse Lymph_r4_dc_0503-0512.indd 510

5/27/2015 12:44:22 PM

Chapter 37  Reverse Lymphatic Mapping

511

R EFERENCES 1. Vignes S, Blanchard M, Yannoutsos A, et al. Complications of autologous lymph-node transplantation for limb lymphoedema. Eur J Vasc Endovasc Surg 45:516-520, 2013. 2. Viitanen TP, Mäki MT, Seppänen MP, et al. Donor-site lymphatic function after microvascular lymph node transfer. Plast Reconstr Surg 130:1246-1253, 2012. 3. Azuma S, Yamamoto T, Koshima I. Donor-site lymphatic function after microvascular lymph node transfer should be followed using indocyanine green lymphography. Plast Reconstr Surg 131:443e-444e, 2013. 4. Viitanen TP, Suominen EA, Saaristo AM. Reply: Donor-site lymphatic function after microvascular lymph node transfer should be followed using indocyanine green lymphography. Plast Reconstr Surg 131:44e, 2013. 5. Pons G, Masia J, Loschi P, et al. A case of donor-site lymphoedema after lymph node-superficial circumflex iliac artery perforator flap transfer. J Plast Reconstr Aesthet Surg 67:119-123, 2014. 6. Becker C, Assouad J, Riquet M, et al. Postmastectomy lymphedema: long-term results following microsurgical lymph node transplantation. Ann Surg 243:313-315, 2006. 7. Lin CH, Ali R, Chen SC, et al. Vascularized groin lymph node transfer using the wrist as a recipient site for management of postmastectomy upper extremity lymphedema. Plast Reconstr Surg 123:12661275, 2009. 8. Althubaiti GA, Crosby MA, Chang DW. Vascularized supraclavicular lymph node transfer for lower extremity lymphedema treatment. Plast Reconstr Surg 131:133e-135e, 2013. 9. Thompson M, Korourian S, Henry-Tillman R, et al. Axillary reverse mapping (ARM): a new concept to identify and enhance lymphatic preservation. Ann Surg Oncol 14:1890-1895, 2007. 10. Klimberg VS. A new concept toward the prevention of lymphedema: axillary reverse mapping. J Surg Oncol 97:563-564, 2008. 11. Dayan JH, Dayan E, Smith ML. Reverse lymphatic mapping: a new technique for maximizing safety in vascularized lymph node transfer. Plast Reconstr Surg 135:277-285, 2015. 12. Dayan JH, Dayan E, Kagen A, et al. The use of magnetic resonance angiography in vascularized groin lymph node transfer: an anatomic study. J Reconstr Microsurg 30:41-45, 2014. 13. Dayan JH, Smith ML, Sultan M, et al. Axillary lymph node transfer for the treatment of lymphedema: technique and case series [abstract]. Plast Reconstr Surg 132(4S-1):86, 2013.

K23349_Neligan_37_Reverse Lymph_r4_dc_0503-0512.indd 511

5/27/2015 12:44:22 PM

K23349_Neligan_37_Reverse Lymph_r4_dc_0503-0512.indd 512

5/27/2015 12:44:22 PM

C hapter 38 Local Flaps and Vascularized Lymph Node Transfer Joshua Levine, Corinne Becker

K ey P oints • The thoracodorsal artery perforator (TDAP) flap and vascularized lymph node transfer (VLNT) are harvested simultaneously with the patient in the decubitus position. • Great care must be taken to preserve the potential recipient vessels in the axilla and lateral chest during the dissection of the TDAP.

Loc

• If no suitable recipient vessels are found, the TDAP pedicle may be used in an end-to-side anastomosis. • Preoperative magnetic resonance angiography or computed tomography angiography is helpful but unnecessary to confirm the viability of the TDAP pedicle.

The treatment for breast cancer can include a wide variety of surgical options, including lumpectomy, partial mastectomy, mastectomy, and any degree of axillary lymph node sampling. These procedures can result in both functional and cosmetic deformities requiring reconstruction. There are various options for autologous breast reconstruction, which are tailored to the specific defect. Reconstruction of the axilla may be indicated if the patient develops lymphedema of the upper extremity, which occurs in up to 42% of patients after lymphatic dissection during surgery for breast cancer.1 The vascularized lymph node transfer (VLNT) is an effective way to treat postoperative lymphedema by reconstructing damaged lymphatic anatomy.2-4 VLNT is a microsurgical free tissue transfer that transplants healthy lymph nodes to the area where lymph nodes have been either removed or traumatized. For patients who have lymphedema and need breast reconstruction, these two reconstructive operations can be combined. Microsurgical (free flap) autologous breast reconstruction with simultaneous lymph node transfer as part of the breast flap has been described.2

513

K23346_Neligan_38_Local Flaps_r4_dc_0513-0518.indd 513

5/26/2015 10:08:30 AM

514

Part V  Current Treatment of Lymphedema

In this chapter I will describe the use of local pedicled perforator flaps for partial breast reconstruction combined with VLNT for axillary reconstruction. Two case examples show how the lateral thoracic flap, specifically the thoracodorsal artery perforator (TDAP), can be combined with simultaneous VLNT to reconstruct two different breast deformities in patients who also have lymphedema.

Mastectomy of the Right Breast With a Failed Expander This 49-year-old woman had hypertension, depression, and obesity (BMI 39.6). She underwent a modified radical mastectomy and axillary lymph node dissection of the right breast for breast cancer, and a submuscular expander was placed for reconstruction. The expander leaked and was never expanded. She presented for right breast reconstruction and also had right arm lymphedema. Her symptoms included right arm swelling (Fig. 38-1, A). Her anatomy was suitable for autologous reconstruction with a lateral thoracic pedicled perforator flap because of the extended dog-ear on her chest from her previous mastectomy. This created an excess of lateral thoracic tissue that could be used for breast reconstruction (Fig. 38-1, B). A CT angiogram was obtained to evaluate the perfusion of the thoracic tissue, and a large TDAP was identified supplying this tissue (Fig. 38-1, C). The patient underwent a TDAP total breast reconstruction with simultaneous VLNT (Fig. 38-1, D). The VLNT was harvested from the patient’s lower abdomen, just cephalad to the right inguinal ligament (Fig. 38-1, E), and was transferred to the right axilla. The recipient vessels were branches of the TDAP pedicle. SPY indocyanine green intraoperative angiography was used to confirm the perfusion of the lymphatic tissue (Fig. 38-1, F). The patient did well after surgery. Her symptoms improved, and she had a reduction in forearm circumference (Fig. 38-1, G).

K23346_Neligan_38_Local Flaps_r4_dc_0513-0518.indd 514

5/26/2015 10:08:30 AM

Chapter 38  Local Flaps and Vascularized Lymph Node Transfer

A

B

D

C

F

515

E

G

FIG. 38-1  A, This patient presented after a mastectomy of the right breast with a failed expander. B, A significant dog-ear from the mastectomy of the right breast was available to be used as a TDAP flap. C, This CT angiogram of the right side of the chest shows a blood vessel going to the TDAP flap. D, The right TDAP flap on its pedicle is shown ready to pass under the anterior edge of the latissimus. E, The right lower abdominal lymph node flap isolated on the superficial epigastric vessels. F, A SPY intraoperative angiogram showing perfusion of the transferred lymph node flap. The numbers represent the level of indocyanine green contrast dye in the perfused tissue. Higher number indicates more perfusion. G, The patient is shown after reconstruction of the right breast with a TDAP flap and simultaneous VLNT. Her symptoms improved, and she had a reduction in forearm circumference.

K23346_Neligan_38_Local Flaps_r4_dc_0513-0518.indd 515

5/26/2015 10:08:31 AM

516

Part V  Current Treatment of Lymphedema

Right Breast Reconstruction With a TDAP Flap and Simultaneous VLNT This 57-year-old woman presented with a history of hypertension and hepatitis C. She had cancer of the left breast, had undergone bilateral mastectomies and left axillary lymph node dissection (ALND) and radiotherapy, resulting in lymphedema of the left arm. Her lymphedema symptoms included frequent and regular episodes of cellulitis requiring hospitalization and intravenous antibiotics. She had extensive liposuction of her abdomen, which rendered the abdomen unavailable for use as a donor site for breast reconstruction (Fig. 38-2, A). She underwent bilateral profunda artery perforator (PAP) flap breast reconstruction and developed an infection and wound healing issues on the left side. This resulted in flap deformity and asymmetry of the left breast (Fig. 38-2, B). She then underwent a left TDAP flap reconstruction of the left breast with simultaneous VLNT to augment the reconstruction. The nodes were transferred from the lower abdomen, and microanastomosis was performed to the TDAP pedicle. The TDAP flap was used to improve the volume and shape of the left breast reconstruction, and the VLNT treated her lymphedema (Fig. 38-2, C and D). Her lymphedema symptoms improved, and she has had no further episodes of infection in the 3 years following the VLNT (Fig. 38-2, E). A comparison of MR lymphatic studies before and after the VLNT shows the clear establishment of new lymph channels leading from the left arm to the chest (Fig. 38-2, F).

K23346_Neligan_38_Local Flaps_r4_dc_0513-0518.indd 516

5/26/2015 10:08:31 AM

Chapter 38  Local Flaps and Vascularized Lymph Node Transfer

A

517

B

C

E

D

F

FIG. 38-2  A, This patient had undergone bilateral mastectomies, left ALND, and radiotherapy, which resulted in left arm lymphedema. B, After bilateral PAP flap reconstruction, she developed a left side deformity as a result of radiotherapy and fat necrosis. C, 37-10 Transfer of the left TDAP flap. D, Harvest of the left lower abdominal lymph node flap. E, The results are shown 3 years postoperatively. F, The postoperative MRI lymphatic study shows that 3 years after the procedure, new lymph channels have been established from the left arm to the chest.

K23346_Neligan_38_Local Flaps_r4_dc_0513-0518.indd 517

5/26/2015 10:08:33 AM

518

Part V  Current Treatment of Lymphedema

Conclusion Autologous lymph node transfer is gaining popularity around the world as the benefits become more established and microsurgeons become more comfortable with the specific supermicrosurgical techniques required to perform them. The increasing use of VLNT will inevitably lead to more creative applications. These two cases show how VLNT can be performed with local flaps, in this case the TDAP.

C linical P earls • VLNT reconstructs damaged lymphatic anatomy and thus is an effective treatment for postoperative lymphedema. • For patients who have lymphedema and need breast reconstruction, these two reconstructive surgeries can be combined. • The lateral thoracic flap, specifically the TDAP, can be performed simultaneously with VLNT to reconstruct various breast deformities in patients with lymphedema.

R EFERENCES 1. Norman SA, Localio AR, Potashnik SL, et al. Lymphedema in breast cancer survivors: incidence, degree, time course, treatment, and symptoms. J Clin Oncol 27:390-397, 2009. 2. Becker C, Assouad J, Riquet M, et al. Postmastectomy lymphedema: long-term results following microsurgical lymph node transplantation. Ann Surg 243:313-315, 2006. 3. Becker C, Vasile JV, Levine JL, et al. Microlymphatic surgery for the treatment of iatrogenic lymphedema. Clin Plast Surg 39:385-398, 2012. 4. Granzow JW, Soderberg JM, Kaji AH, et al. Review of current surgical treatments for lymphedema. Ann Surg Oncol 21:1195-1201, 2014.

K23346_Neligan_38_Local Flaps_r4_dc_0513-0518.indd 518

5/26/2015 10:08:33 AM

C hapter 39 Transverse Myocutaneous Gracilis With Vascularized Lymph Node Transfer Sinikka Suominen, Maija Kolehmainen

K ey P oints • Breast reconstruction and simultaneous vascularized lymph node transfer can be done with a gracilis flap and its transverse skin island from the upper thigh. • A second vascular anastomosis may be necessary to the lymph node part of the flap.

The combination of free vascularized lymph node transfer (VLNT) with breast reconstruction seems to be the optimal treatment for a postmastectomy patient who has postoperative lymphedema of the upper extremity and wants breast reconstruction. Abdominal flaps (transverse rectus abdominis myocutaneous flap, deep inferior epigastric perforator [DIEP] flap, and superficial inguinal epigastric artery [SIEA] flap) are the benchmark for breast reconstruction, and inguinal lymph nodes can conveniently be included in these flaps.

Tra Lym

However, not all mastectomy patients are candidates for abdominal flap transfer. Lower abdominal tissue volume may be inadequate because of slimness or body type, and multiple or midline scars from previous abdominal surgery can hinder the use of that area. Quite often even a slim young patient has a useful donor site in the upper thigh, and because of the inconspicuous placement of the scar, many patients prefer it over the donor site of the latissimus dorsi or thoracodorsal artery perforator flap in the back. In our institution we have offered the transverse upper gracilis flap since 2006 as one choice for breast reconstruction, patient anatomy permitting. The use of the upper thigh as a donor site was first described by Yousif et al1 in 1992, and the first suggestion for its use in breast reconstruction in the English-speaking literature was by Arnez et al2 in 2002, who called it the transverse upper gracilis flap. Actually, Peek et al3 in Germany had already described the use of the same skin island in breast reconstruction in 2002, but as a perforator-based flap. Schoeller et al4 popularized this method and renamed the flap transverse myocutaneous gracilis (TMG). 519

K23346_Neligan_39_Transverse_r3_dc_0519-0526.indd 519

5/26/2015 10:09:44 AM

520

Part V  Current Treatment of Lymphedema

In 2010 we performed the first combined TMG and VLNT on a patient with small A cup breasts in whom we could not use the lower abdomen as a donor site. Because we already performed DIEP and SIEA flaps with a VLNT routinely and obtained similar favorable outcomes as reported in the literature, it seemed natural to try to combine these procedures with another anatomically linked donor site. Because these patients have two problems, the lack of a breast and lymphedema, they seek help for both.

Preoperative Assessment The TMG is planned as usual, but the drawing of the skin incision continues over the inguinal crease and either along the iliac spine linking the TMG flap to a groin flap or along the hairline and SIEA vessels. The markings are first done with the patient standing, marking the lateral edge of the pubic hair line, the inguinal ligament, and the femoral vessels. As in any TMG flap, the width of the skin island in the thigh is determined by pinch grip, and the upper edge of the island is placed as high as possible. In the back, the midline of the buttocks and the lateral tip of the gluteal fold are marked, because the skin island should not exceed that point to stay invisible. The patient is then placed in the supine position. The femoral vessels are palpated, and the SIEA and superficial circumflex iliac artery (SCIA) vessels are identified with pencil Doppler ultrasound and are marked on the skin. The skin island of the groin and superficial circumflex iliac perforator flaps is designed to extend at least 5 to 8 cm from the estimated position of the lymph nodes to serve as a deepithelialized bridge crossing the scarred axilla and also to allow the use of a monitoring skin island. The skin island is positioned according to the Doppler signal and can either be more vertical on the SIEA or more horizontal on the SCIA vessels. Either a contralateral or an ipsilateral flap can be designed according to the surgeon’s preference. We usually use an ipsilateral flap for easier mobilization of the patient. With a gynecologic table, both sides are easy to harvest with a two-team approach: one team prepares the recipient site and releases the axillary scar at the same time the flap is harvested.

Surgical Technique We prefer to start by raising the vascularized lymph node flap as described by Becker and Hidden,5 Chen et al,6 and Becker et al7 (see Chapter 38). The skin island is most easily raised from lateral to medial and superior to inferior. The lateral part of the skin island can be raised above Scarpa’s fascia as a superficial circumflex iliac perforator flap, but because the lymph nodes derive their vascularity from the actual source vessel, the surgeon should follow the vessels and work one’s way deeper at the level of the fascia and eventually below it to expose the femoral vessels and origin of the pedicle. This is an area of vast anatomic variability, and either the SIEA or the SCIA vessels can be used to carry the vascularized lymph nodes. The literature warns about going below the level of the inguinal ligament to avoid injuring the lymph drainage of the lower extremity8,9 (see

K23346_Neligan_39_Transverse_r3_dc_0519-0526.indd 520

5/26/2015 10:09:44 AM

Chapter 39  Transverse Myocutaneous Gracilis With Vascularized Lymph Node Transfer

521

A

B

FIG. 39-1  A, The skin of the flap should be incised, because posterior markings tend to disappear. B, Raising the lymph node flap.

Chapter 5). It is advisable, especially at the beginning of a surgeon’s experience, to use reversed mapping to monitor that all nodes draining the lower extremity are excluded. Patent blue injected to the distal tip of the flap may also be helpful. The skin island of the groin and vascularized lymph node flap is designed to join the skin island of the TMG and should be kept as wide as the skin closure allows. As soon as the decision is made regarding which lymph nodes will be included in the flap, the dissection of the flap continues medially at a level immediately below Scarpa’s fascia until medial to the saphenous vessels (Fig. 39-1, A). The critical lymph vessels of the lower extremity usually lie below the inguinal ligament medial to the femoral vein and are not touched. The surgeon can again go deep to the level of the fascia and continue to raise the TMG flap as usual. We usually keep the pedicle of the lymph node flap connected, and it is divided only after the whole flap has been raised at the same time as the actual gracilis pedicle (Fig. 39-1, B). The TMG is raised in a standard fashion, identifying the saphenous vein and dividing its branches to the skin island. The fat is raised at the level of the fascia over the adductor longus muscle, and blunt dissection of the loose areolar space between the adductor and gracilis reveals the pedicle. After the pedicle has been identified, it is easy to proceed distally along the gracilis muscle. This can usually be done bluntly, with the fingers distally, up to the tendinous insertion of the muscle. The secondary pedicle is clipped and divided. Posterior to the gracilis, the flap is raised in a suprafascial plane, tapering the subcutaneous tissue 2 to 5 cm below the distal skin edge to recruit as much volume as possible. After the muscle has been divided and the posterior part of the flap released, the surgeon dissects the vascular pedicle, ligating and dividing the branches to the adductor longus and sharply dividing the motor nerve to the gracilis. If necessary, the proximal part of the pedicle can also be exposed from lateral to the adductor longus muscle.

K23346_Neligan_39_Transverse_r3_dc_0519-0526.indd 521

5/26/2015 10:09:45 AM

522

Part V  Current Treatment of Lymphedema

FIG. 39-2  TMG flap with vascularized lymph nodes.

A

B

FIG. 39-3  A, Flap positioned on the chest. B, Lymph node flap deepithelialized.

Before the pedicle is divided, the surgeon should temporarily occlude the pedicle of the lymph node part of the flap to evaluate the vascularity of the nodes on the gracilis pedicle alone (Fig. 39-2). In our clinical experience the flap bled well in two out of three patients, and a second anastomosis was unnecessary. Perioperative indocyanine green angiography is a helpful aid in this decision-making. As in any VLNT, meticulous release of the axillary scar is necessary, and a space is created along the axillary vein in the proximal arm. The tip of the deepithelialized skin island is placed in this pocket and temporarily fixed through the skin (Fig. 39-3). The nodes are placed along the axillary vessels and a breast is created (Fig. 39-4). When TMG is combined with vascularized lymph nodes, we prefer to use the thoracodorsal or circumflex scapular vessels as the recipient vessels. When the thoracodorsal vessels are divided at a level above the serratus branch, they can easily be transposed to the pectoralis margin and anastomosed to the gracilis pedicle without tension. Because the scar at the donor site has a tendency to broaden, the surgeon should use some fixation sutures, such as in a medial thigh lift procedure. The skin is often closed with barbed sutures, and two suction drains are used—one in the thigh and one in the groin.

K23346_Neligan_39_Transverse_r3_dc_0519-0526.indd 522

5/26/2015 10:09:45 AM

Chapter 39  Transverse Myocutaneous Gracilis With Vascularized Lymph Node Transfer

523

FIG. 39-4  The finished breast.

Postoperative Care Compression garments or bandages are useful in the immediate postoperative period. The patients are mobilized with the aid of a physical therapist on the first postoperative day but are advised to avoid full lateral rotation or abduction of the hip for the first weeks. Normal physical activities and sports can resume in 6 weeks. To monitor any possible donor-site problems, it is advisable to measure the circumference or volume of the lower extremities postoperatively and at 1 year.9,10

Complications and Patient Outcomes Because the secondary pedicle of the gracilis is divided and ligated with limited visibility, a clip may loosen and result in significant bleeding at the donor site. To avoid this, the surgeon should take extra care in ligating this vessel and also in using postoperative compression bandaging. A seroma of the inguinal donor site is not uncommon, and prolonged drainage for 5 to 7 days or repeated punctures at postoperative controls may be necessary. The patient should be informed regarding this possibility before surgery. Lymphedema of the donor site is the most feared complication, and Pons et al10 reported one case of lymphedema in a series of 42 patients with VLNT. Viitanen et al9 reported subclinical temporary impairment of lymph flow after combined DIEP and lymph node transfer. In our own unpublished series of seven patients with TMG and VLNT, one patient developed transient minor clinical lymphedema of the donor leg. Although her lymphoscintigraphic findings showed diminished flow at 1 year, this resolved and was again normal 2 and 3 years after surgery. At our institution, we routinely follow all patients postoperatively up to 3 years, after which the follow-up continues by the general practitioner or a therapist. However, if any unexpected changes appear, patients can request an appointment at any time. Patient outcomes are similar to the transfer of vascularized lymph nodes with a groin flap or together with an abdominal flap.7,8,11,12 Strong experimental data support this technique, although

K23346_Neligan_39_Transverse_r3_dc_0519-0526.indd 523

5/26/2015 10:09:46 AM

524

Part V  Current Treatment of Lymphedema

the exact mechanism of the restoration or enhancement of lymphatic function is still unclear.13,14 The breast volume that can be achieved from the thigh is not as much as from an abdominal flap. Typically volumes of 300 to 600 cc can be harvested, but this donor site can usually be used even in thin women who have no fat in the abdomen. Atrophy of the muscle may result in diminishing of the final volume by 1 year, and complimentary touch-up fat grafting may be necessary as with any breast reconstruction method.

Conclusion The combination of breast reconstruction with the TMG flap and free VLNT is a possible solution in patients in whom abdominal flaps cannot be used. However, this procedure carries the risk of a donor-site problem, and the patient must be well informed and carefully followed. Meticulous technique and experience in raising vascularized lymph node flaps are of utmost importance to avoid the danger zones. In experienced hands it is a useful procedure that allows the solution of two problems at once, the lymphedema and a missing breast.

C linical P earls • TMG is ideal for bilateral reconstruction; a gynecologic table allows a three-team approach. • In large-volume thighs there is no need to include the whole muscle; it can be cut at the level of the skin island. • Reverse sentinel mapping helps to exclude critical lower extremity nodes.

R EFERENCES 1. Yousif NJ, Matloub HS, Kolachalam R, et al. The transverse gracilis musculocutaneous flap. Ann Plast Surg 29:482-490, 1992. 2. Arnez ZM, Pogorelec D, Planinsek F, et al. Breast reconstruction by the free transverse gracilis (TUG) flap. Br J Plast Surg 57:20-26, 2004. 3. Peek A, Müller M, Exner K. [The free gracilis perforator flap for autologous breast reconstruction] Handchir Mikrochir Plast Chir 34:245-250, 2002. 4. Schoeller T, Huemer GM, Wechselberger G. The transverse musculocutaneous gracilis flap for breast reconstruction: guidelines for flap and patient selection. Plast Reconstr Surg 122:29-38, 2008. 5. Becker C, Hidden G. [Transfer of free lymphatic flaps. Microsurgery and anatomical study] J Mal Vasc 13:119-122, 1988. 6. Chen HC, O’Brien BM, Rogers IW, et al. Lymph node transfer for the treatment of obstructive lymphoedema in the canine model. Br J Plast Surg 43:578-586, 1990. 7. Becker C, Assouad J, Riquet M, et al. Postmastectomy lymphedema: long-term results following microsurgical lymph node transplantation. Ann Surg 243:313-315, 2006. 8. Saaristo AM, Niemi TS, Viitanen TP, et al. Microvascular breast reconstruction and lymph node transfer for postmastectomy lymphedema patients. Ann Surg 255:468-473, 2012.

K23346_Neligan_39_Transverse_r3_dc_0519-0526.indd 524

5/26/2015 10:09:46 AM

Chapter 39  Transverse Myocutaneous Gracilis With Vascularized Lymph Node Transfer

525

9. Viitanen TP, Maki MT, Seppanen MP, et al. Donor-site lymphatic function after microvascular lymph node transfer. Plast Reconstr Surg 130:1246-1253, 2012. 10. Pons G, Masia J, Loschi P, et al. A case of donor-site lymphoedema after lymph node-superficial circumflex iliac artery perforator flap transfer. J Plast Reconstr Aesthet Surg 67:119-123, 2014. 11. Raju A, Chang DW. Vascularized lymph node transfer for treatment of lymphedema: a comprehensive literature review. Ann Surg. 2014 Jun 19. [Epub ahead of print] 12. Dancey A, Nassimizadeh A, Nassimizadeh M, et al. A chimeric vascularised groin lymph node flap and DIEP flap for the management of lymphoedema secondary to breast cancer. J Plast Reconstr Aesthet Surg 66:735-737, 2013. 13. Tammela T, Saaristo A, Holopainen T, et al. Therapeutic differentiation and maturation of lymphatic vessels after lymph node dissection and transplantation. Nat Med 13:1458-1466, 2007. 14. Shesol BF, Nakashima R, Alavi A, et al. Successful lymph node transplantation in rats, with restoration of lymphatic function. Plast Reconstr Surg 63:817-823, 1979.

K23346_Neligan_39_Transverse_r3_dc_0519-0526.indd 525

5/26/2015 10:09:46 AM

K23346_Neligan_39_Transverse_r3_dc_0519-0526.indd 526

5/26/2015 10:09:46 AM

C hapter 40 Lymphatic Microsurgical Preventing Healing Approach Concept Francesco Boccardo

Lym Con

K ey P oints • The lymphatic microsurgical preventing healing approach (LYMPHA) is a surgical technique for the primary prevention of arm lymphedema in patients with breast cancer, although it can also be used in patients with melanoma localized to the trunk. • The learning curve for the LYMPHA procedure is approximately 30 cases. • Lymphovenous shunts are used in the LYMPHA technique because they are faster, more physiological, and do not require lymph node or lymph vessel harvesting. • Axillary reverse mapping (ARM), which uses a small amount of blue dye, was developed to identify the lymphatic pathways draining the arm. • Lymphaticovenous anastomosis prevents lymphedema and reduces early lymphatic complications resulting from reduced regional intralymphatic pressure.

A side effect of axillary lymph node excision and radiotherapy for breast cancer is arm lymphedema in about 25% of patients (range 13% to 52%). Sentinel lymph node (SLN) biopsy has reduced the severity of swelling to nearly 6% (from 2% to 7%), but in a patient with a positive SLN, complete axillary dissection is still required. This is why the axillary reverse mapping (ARM) method1,2 was developed—to identify and preserve the lymphatics draining the arm. The procedure consists of an intradermal and a subcutaneous injection of a small quantity (1 to 2 ml) of blue dye at the medial surface of the arm, which helps locate the lymphatic pathways draining the arm. The ARM technique assists in identifying the variable clinical anatomy that differs from what is already generally appreciated, that is, the most common location of arm lymphatics below and This chapter was adapted from Boccardo F, Casabona F, De Cian F, Friedman D, Murelli F, Puglisi M, Campisi C, Molinari L, Spinaci S, Dessalvi S, Campisi C. Lymphatic Microsurgical Preventing Healing Approach (LYMPHA) for primary surgical prevention of breast cancer-related lymphedema: over 4 years follow-up. Microsurgery 34:421-424, 2014; with permission from the publisher.

527

K23349_Neligan_40_Lymph Microsurg_r3_dc_0527-0538.indd 527

5/27/2015 12:46:02 PM

528

Part V  Current Treatment of Lymphedema

around the axillary vein. In about one third of patients, blue lymphatics extended to within 3 to 4 cm of the vein, the site at which the SLNs were easily located. This explained the occurrence of lymphedema after only SLN biopsy.3 The ARM procedure also showed that blue nodes were almost always placed at the lateral part of the axilla, under the vein, and above the second intercostobrachial nerve.4 The risk of arm lymphedema was decreased if the lymph nodes related to arm lymphatic drainage remained in place, but the main risk of not retrieving all the nodes was leaving metastatic disease in the axilla. On the contrary, the arm lymphatic pathways when they enter the axilla intuitively cannot be the site of a metastatic breast tumor, and their preservation certainly brings about a significant decrease in the incidence of lymphedema. Based on long-term experience with lymphaticovenous anastomosis5,6 for lymphedema treatment, we developed and performed preventive lymphaticovenous anastomosis during nodal dissection. We7 reported on the long-term outcome of our experience in the prevention of breast cancer– related lymphedema with the lymphatic microsurgical preventing healing approach (LYMPHA), which consisted of anastomosis of the arm lymphatics to a collateral branch of the axillary vein.

Clinical Experience From July 2008 to December 2012, 78 patients underwent axillary nodal dissection for breast cancer treatment, 74 of whom underwent the LYMPHA procedure (Table 40-1). The LYMPHA procedure could not be performed in four patients because no afferent lymphatics could be found (three patients) and because of massive metastatic disease (one patient). The indications for the LYMPHA technique were based on clinical and lymphoscintigraphic parameters (Fig. 40-1). It is well known that obesity predisposes an individual to lymphedema, and therefore patients with a BMI higher than 30 were considered at high risk for lymphedema and candidates for LYMPHA. Patients with a normal BMI were studied with lymphoscintigraphy, which indicated latent lymphatic impairment still not evident clinically.

TABLE 40-1  Patient Characteristics Variable

Patient Group

Age (yr)

57 (42-69)

BMI

24 (21-33)

Number of lymph nodes retrieved

19 (12-21)

Number of metastatic lymph nodes

3 (0-4)

Type of surgery (mastectomy/tumorectomy cases)

34/40

Radiotherapy

35 underwent radiotherapy; 39 did not

Cellulitis

14 had cellulitis; 60 did not

K23349_Neligan_40_Lymph Microsurg_r3_dc_0527-0538.indd 528

5/27/2015 12:46:02 PM

Chapter 40  Lymphatic Microsurgical Preventing Healing Approach Concept

529

CLINICAL CRITERIA BMI

,30

$30 LYMPHA

INSTRUMENTAL CRITERIA TI ,10

LS (TI)

TI $10 LYMPHA

FIG. 40-1  Clinical and instrumental selection parameters for the use of the LYMPHA technique. (LS, Lymphoscintigraphy; TI, transport index.)

Venous branch

LVA

LVA

Blue lymphatics

Blue dye Blue lymphatics

FIG. 40-2  The LYMPHA technique at the left axilla after lymph nodal dissection for breast cancer. (LVA, Lymphaticovenous anastomosis.

Regarding lymphatic arm drainage, blue nodes were identified in almost all patients after blue dye injection at the medial surface of the arm. All blue nodes were resected, and two to four main afferent lymphatics from the arm were prepared and used for anastomoses. Lymphatics were introduced inside the vein cut-end by a U-shaped stitch. Several other stitches were inserted to fix the lymphatic adventitia to the vein wall (Fig. 40-2). The operation usually took only 20 minutes on average, because both the lymphatics and vein were prepared during nodal dissection. Drain tubes were removed after about 10 days (range 7 to 12 days). No bandages were necessary on the arm, and patients did not wear any elastic garment. Volumetry was performed preoperatively in all patients and after 1, 3, 6, and 12 months and once a year. Lymphoscintigraphy was performed in 45 patients preoperatively and also in 30 patients postoperatively after at least 1 year, comparing the preoperative and postoperative lymph transport indexes.8 Statistical analysis was performed by Fisher’s exact test (p ,0.05).

K23349_Neligan_40_Lymph Microsurg_r3_dc_0527-0538.indd 529

5/27/2015 12:46:03 PM

530

Part V  Current Treatment of Lymphedema

Long-Term Outcome Seventy-one patients had no sign of lymphedema, and volumetric measurements were the same as before surgery (Table 40-2). In three patients from the initial clinical experience, lymphedema occurred 8 to 12 months after surgery, usually with the appearance of lymphangitic attacks. Radiotherapy caused temporary edema of the arm in eight patients, and the edema became permanent in three patients. Thus the incidence of secondary arm lymphedema after the LYMPHA technique was 4.05%. Lymphoscintigraphy showed the patency of the lymphaticovenous anastomosis at 1 to 4 years after surgery. When we compared the lymphatic transport index with preoperative conditions, the results showed a significant improvement in all patients with early liver tracer uptake (Fig. 40-3). Lymphoscintigraphy was always performed in the same department, which studied the superficial lymphatic circulation by injecting the tracer into the dorsum of the hand.

TABLE 40-2  Clinical and Instrumental Assessment of Results After the LYMPHA Procedure in 74 Patients Volumetry (ml)

Lymphoscintigraphy Transport Index

Postoperative

Postoperative

Number of Patients

Preoperative

1 yr

4 yr

Preoperative

1 yr

4 yr

71

765.7 6 88.7

763.4 6 97.6

761.9 6 89.6

12 6 3.7

9 6 4.02

8 6 3.8

3

773.5 6 120.8

1035.7 6 20.8



14 6 4.1

16 6 3.7



p ,0.05 preoperative and postoperative volumetric measurements in 71 patients.

A A

C

B

K23349_Neligan_40_Lymph Microsurg_r3_dc_0527-0538.indd 530

FIG. 40-3  A, Preoperative and B, postoperative lymphoscintigraphy after left axillary dissection and preventive lymphaticovenous anastomosis. Arrows indicate that a good lymphatic pathway appeared after axillary nodal dissection and LYMPHA technique. C, Liver uptake.

5/27/2015 12:46:03 PM

Chapter 40  Lymphatic Microsurgical Preventing Healing Approach Concept

531

Potential for Applying the LYMPHA Concept to Melanoma Patients Lymphaticovenous anastomosis not only prevented lymphedema, but also reduced early lymphatic complications (for example, lymphorrhea and lymphocele) because of the reduced regional intralymphatic pressure.7 In the axilla, new lymphatic vessel formation (lymphangiogenesis) occurred in response to the ligation of lymphatic vessels involved in lymph node retrieval. Lymphangiogenesis and lymphatic hypertension were found experimentally in lymphatic drainage obstruction. In response to lymphatic hypertension, lymphovenous shunts opened and provided alternative lymphatic pathways when the main ones were obstructed. These mechanisms represented an adaptive response to lymphatic hypertension but were not enough to restore normal flow parameters.9,10 Furthermore, chronic obstruction to lymph flow progressively led to reduced lymphatic contractility, lymphatic thrombosis, and fibrotic changes at a different degree according to variable constitutional predisposition.9,10 Recent advances in the treatment of breast cancer, specifically related to the prevention of lymphatic complications after SLN biopsy and axillary dissection, led to the proposal of a new technique to primarily prevent lymphedema by microsurgical lymphaticovenous anastomosis (LYMPHA).7 Lymphovenous shunts11 rather than lymph node or lymph vessel transplantation were chosen for the LYMPHA technique because it is a faster technique and more physiological and does not require lymph node or lymph vessel harvesting.12 The ARM technique4 facilitated the identification of arm lymphatics and lymph nodes that could be preserved, even though there was a theoretic risk of leaving undetected metastatic disease in the axilla. However, it was almost impossible to preserve efferent lymphatics from the blue nodes, because they joined the common axillary nodal basin draining the breast. Thus the inability to preserve efferent lymphatics made it practically impossible to preserve arm lymphatic flow. Therefore based on our wide experience in the treatment of lymphedema with microsurgical lymphaticovenous anastomosis, we performed lymphaticovenous anastomosis immediately after completion of nodal axillary excision. The surgical technique proposed for patients with operable breast cancer requiring an axillary dissection consisted of lymphaticovenous anastomosis between the arm lymphatics that were simultaneously identified by the injection of blue dye in the arm and an axillary vein branch (Fig. 40-4). It was almost always possible to find blue lymphatics and also a vein branch long enough to connect to the arm lymphatics, which were usually located very laterally (Fig. 40-5). The disruption of the axillary nodes and closure of the arm lymphatics explained the significantly high risk of early and late lymphatic complications after axillary dissection. The most serious complication was arm lymphedema.13 Therefore the incidence of secondary arm lymphedema after the LYMPHA technique was 4.05% compared with the incidence after complete axillary dissection reported in the literature, which ranged from 13% to 65%, depending on the criteria used and means of assessment.9,14,15 Finally, it is important to underscore that all three patients who developed lymphedema after the LYMPHA technique were from the preliminary clinical experience. Thus we conclude that the learning curve for LYMPHA is about 30 cases.

K23349_Neligan_40_Lymph Microsurg_r3_dc_0527-0538.indd 531

5/27/2015 12:46:03 PM

532

Part V  Current Treatment of Lymphedema

Blue nodes related to arm drainage identified

Axillary vein

Injection site Lymphatic vessels draining arm

Breast tumor and affected nodes removed

Collateral venous branch Intercostobrachial nerve Axillary lymph nodes Lymphatic vessels draining breast tissue Tumor

Arm vessels joined to collateral venous branch using lymphaticovenous anastomosis

FIG. 40-4  The LYMPHA technique at the arm.

FIG. 40-5  Surgical phase of the LYMPHA technique at the axilla.

K23349_Neligan_40_Lymph Microsurg_r3_dc_0527-0538.indd 532

5/27/2015 12:46:03 PM

Chapter 40  Lymphatic Microsurgical Preventing Healing Approach Concept

533

Use of LYMPHA to Prevent Lymphedema After Treatment for Melanoma The LYMPHA technique is also useful in patients with melanoma of the trunk, in whom it is possible to perform preventive lymphaticovenous anastomosis simultaneous with inguinal lymphadenectomy (Figs. 40-6 and 40-7). The blue dye is injected at the medium third of the medial surface of the thigh. Blue lymphatics afferent to the groin are isolated and anastomosed to a collateral branch of the great saphenous vein. In most cases, the medial accessory saphenous vein is used, and all blue afferent lymphatics are anastomosed to the same vein segment (multiple lymphaticovenous anastomoses). A telescopic technique (several lymphatics introduced into the venous segment) is used. This procedure permits the lymph coming from the main lymphatic pathways of the limb to flow into the bloodstream. This preventive surgical approach can only be performed in patients with melanoma localized to the trunk (Fig. 40-8). In melanoma of the extremities, this preventive approach cannot be used for oncologic reasons, and lymphedema development can be limited with fastidious postoperative management. Immediate postoperative elastic wraps have proved their efficacy when placed in multiple layers from the foot to above the knee and replaced afterward with compression stockings. Leg elevation is also useful when the patient is still lying on the bed. After an accurate oncologic assessment and the absence of any sign of recurrence of the pathology, some cases of lymphedema could be treated subsequently by lymphatic microsurgery to bypass the obstacle to lymph flow by derivative multiple lymphaticovenous anastomoses at the groin.7 In these patients, lymphoscintigraphy was the procedure of choice to assess lymphatic transport capacity before surgery and afterward to evaluate long-term results.5 Venous ultrasonography was performed in all patients to study vein circulation. Above all, lymphaticovenous anastomosis proved successful in the early stages of lymphedema because of the absence of fibrotic changes of tissues and the lymphatic wall. A correctly performed complete lymphadenectomy with careful attention to the surgical detail can help minimize wound complications, decrease the time of closed-suction drains, and decrease venous thromboembolism. Some patients have a predisposition to lymphedema, and even though the etiologic factor seems to be surgical intervention, this may represent the final straw that unearthed a previously unrecognized abnormal lymphatic system. Patients at risk may be assessed preoperatively with lymphoscintigraphy, with the appropriate preventive measures adopted.

K23349_Neligan_40_Lymph Microsurg_r3_dc_0527-0538.indd 533

5/27/2015 12:46:03 PM

534

Part V  Current Treatment of Lymphedema

A

Inguinal lymph node

B

Blue lower limb lymphatics

FIG. 40-6  The LYMPHA technique at the groin. A, Lymphadenectomy. B, Lymphaticovenous anastomosis.

Melanoma and affected nodes removed

Melanoma Lymphatic vessels draining the trunk wall

Inguinal lymph nodes Great saphenous vein

Blue nodes related to leg drainage identified

Collateral venous branch

Lymphatic vessels draining the leg Leg vessels joined to collateral venous branch using lymphaticovenous anastomosis (shirtsleeve)

Injection site

FIG. 40-7  The LYMPHA technique at the lower limb.

K23349_Neligan_40_Lymph Microsurg_r3_dc_0527-0538.indd 534

5/27/2015 12:46:04 PM

Chapter 40  Lymphatic Microsurgical Preventing Healing Approach Concept

535

PREOPERATIVE LYMPHOSCINTIGRAPHY

Anterior

Posterior

Anterior

Posterior

Anterior

Posterior

Anterior

Posterior

Anterior

Posterior

Anterior

Posterior

POSTOPERATIVE LYMPHOSCINTIGRAPHY

Anterior

Posterior

Anterior

Posterior

FIG. 40-8  Preoperative and postoperative lymphoscintigraphic control showing the disappearance of dermal backflow, which was present preoperatively (arrows), and the appearance of a good lymphatic pathway, which was not present preoperatively (asterisk).

K23349_Neligan_40_Lymph Microsurg_r3_dc_0527-0538.indd 535

5/27/2015 12:46:04 PM

536

Part V  Current Treatment of Lymphedema

Conclusion Lymphedema is a consequence of cancer treatment, and patients should be informed of the signs and symptoms and the possibility of preventing lymphedema.16,17 The use of blue dye and lymphaticovenous anastomosis helps solve the problem of preventing secondary arm and leg lymphedema while maintaining oncologic radicality. Therefore LYMPHA may represent a rational approach to the prevention of lymphedema and reduce other lymphatic complications after axillary and groin surgery in the treatment of breast cancer, melanoma, and other tumors.

C linical P earls • Lymphedema is a frequent consequence of cancer treatment, of which patients should be informed. • The use of blue dye and lymphaticovenous anastomosis (LYMPHA technique) can prevent the appearance of secondary lymphedema of the arm and leg. • LYMPHA can offer patients a better quality of life. • LYMPHA should be included in every cancer treatment unit because of the high incidence of lymphedema. • LYMPHA can be easily and rapidly performed with a telescopic technique by anastomosing several lymphatics altogether into the venous segment.

R EFERENCES 1. Thompson M, Korourian S, Henry-Tillman R, et al. Axillary reverse mapping (ARM): a new concept to identify and enhance lymphatic preservation. Ann Surg Oncol 14:1890-1895, 2007. 2. Sakorafas GH, Perosa G, Cataliotti L, et al. Lymphedema following axillary lymph node dissection for breast cancer. Surg Oncol 15:153-165, 2006. 3. Nos C, Lesieur B, Clough KB, et al. Blue dye injection in the arm in order to conserve the lymphatic drainage of the arm in breast cancer patients requiring an axillary dissection. Ann Surg Oncol 14:24902496, 2007. 4. Ponzone R, Mininanni P, Cassina E, et al. Axillary reverse mapping in breast cancer: can we spare what we find? Ann Surg Oncol 15:390-391, 2008. 5. Campisi C, Bellini C, Campisi CC, et al. Microsurgery for lymphedema: clinical research and longterm results. Microsurgery 30:256-260, 2010. 6. Auba C, Marre D, Rodriguez-Losada G, et al. Lymphaticovenular anastomoses for lymphedema treatment: 18 months postoperative outcomes. Microsurgery 32:261-268, 2012. 7. Boccardo F, Casabona F, De Cian F, et al. Lymphedema microsurgical preventive healing approach: a new technique for primary prevention of arm lymphedema after mastectomy. Ann Surg Oncol 16:703708, 2009. 8. Kleinhans E, Baumeister RG, Hahn D, et al. Evaluation of transport kinetics in lymphoscintigraphy: follow-up study in patients with transplanted lymphatic vessels. Eur J Nucl Med 10:349-352, 1985.

K23349_Neligan_40_Lymph Microsurg_r3_dc_0527-0538.indd 536

5/27/2015 12:46:04 PM

Chapter 40  Lymphatic Microsurgical Preventing Healing Approach Concept

537

9. Gärtner R, Jensen MB, Kronborg L, et al. Self-reported arm-lymphedema and functional impairment after breast cancer treatment—a nationwide study of prevalence and associated factors. Breast 19:506515, 2010. 10. Jila A, Kim H, Nguyen VP, et al. Lymphangiogenesis following obstruction of large postnodal lymphatics in sheep. Microvasc Res 73:214-223, 2007. 11. Boccardo F, Fulcheri E, Villa G, et al. Lymphatic microsurgery to treat lympedema: techniques and indication for better results. Ann Plast Surg 7:191-195, 2013. 12. Felmerer G, Sattler T, Lohrmann C, et al. Treatment of various secondary lymphedemas by microsurgical lymph vessel transplantation. Microsurgery 32:171-177, 2012. 13. Francissen CM, Dings PJM, van Dalen T, et al. Axillary recurrence after a tumor-positive sentinel lymph node biopsy without axillary treatment: a review of the literature. Ann Surg Oncol 19:4140-4149, 2012. 14. Deutsch M, Land S, Begovic M, et al. The incidence of arm edema in women with breast cancer randomized on the National Surgical Adjuvant Breast and Bowel Project study B-04 to radical mastectomy versus total mastectomy and radiotherapy versus total mastectomy alone. Int J Radiat Oncol Biol Phys 70:1020-1024, 2008. 15. Kwan ML, Darbinian J, Schmitz KH, et al. Risk factors for lymphedema in a prospective breast cancer survivorship study: the Pathways Study. Arch Surg 145:1055-1063, 2010. 16. Paskett ED, Dean JA, Oliveri JM, et al. Cancer-related lymphedema risk factors, diagnosis, treatment, and impact: a review. J Clin Oncol 30:3726-3733, 2012. 17. Witte MH, Dellinger MT, Papendieck CM, et al. Overlapping biomarkers, pathways, processes and syndromes in lymphatic development, growth and neoplasia. Clin Exp Metastasis 29:707-727, 2012.

K23349_Neligan_40_Lymph Microsurg_r3_dc_0527-0538.indd 537

5/27/2015 12:46:04 PM

K23349_Neligan_40_Lymph Microsurg_r3_dc_0527-0538.indd 538

5/27/2015 12:46:04 PM

C hapter 41 Combined Surgical Treatment for Breast Cancer–Related Lymphedema Jaume Masia

Com Lym

K ey P oints • Accurate diagnosis is essential. • A careful history as well as appropriate imaging helps establish the diagnosis. • Only patients with functioning lymphatic channels are candidates for reconstruction. • Patients with advanced lymphedema are better candidates for excisional procedures.

Breast cancer–related lymphedema (BCRL) is a persistent and debilitating complication that affects the health-related quality of life of breast cancer patients.1-6 Findings from recent studies suggest that as many as 30% of women will develop this condition after breast cancer treatment.7-9 Treatment has traditionally been based exclusively on conservative physical and medical therapies, but surgical approaches have emerged over the years in an attempt to provide a more effective or complementary treatment. Most such surgical approaches can be considered either palliative or reconstructive. Palliative approaches can be excisional or reductive. One of the first techniques described was radical excision, the Charles procedure,10 described in 1912. It consisted of resecting the skin and subcutaneous tissue and applying a skin graft. Results were poor, and this technique is now indicated only in extreme cases of functional limitation. More recently, vibroliposuction (Brorson’s technique) has been used to reduce hypertrophic adipose and fibrotic tissue, and it has been found to improve the quality of life in affected patients.11 Reconstructive approaches aim to improve or restore the functionality of an impaired lymphatic system. Those most commonly used to date are autologous vascularized lymph node transfer (ALNT),12 lymphaticovenular anastomosis (LVA),13-16 and lympholymphatic bypass.17 Because of a lack of consensus regarding the most effective surgical procedure for lymphedema,18 in 2005 539

K23349_Neligan_41_Combined Treat_r2_dc_0539-0552.indd 539

5/27/2015 12:48:06 PM

540

Part V  Current Treatment of Lymphedema

we chose to perform autologous lymph-node transplantation (ALNT), following Becker’s technique,12 and LVA following Koshima’s technique.13 We chose these techniques as we had confidence in their reproducibility and in their rationale.12-14 We did not perform lympholymphatic bypass (Baumeister’s17 technique) because we considered the risk to develop secondary lymphedema and the scarring and morbidity at the donor site was considerable. On the basis of our results using these two approaches, we realized that an accurate preoperative assessment was needed to select the most suitable approach for each individual patient. Our combined surgical treatment evolved from this concept and we have followed these techniques since 2009. To perform preoperative assessment we rely on imaging techniques such as indocyanine green (ICG) lymphography16 and MR lymphography.19,20 Both these techniques are effective for this purpose. Our surgical approach varies, depending on the results of the preoperative study. In patients who have residual function of their lymphatic system, we perform LVA or LVA in combination with ALNT. In later stages of lymphedema, characterized by excess adipose and fibrotic tissue, we opt for a reductive technique (vibroliposuction11). The risk of developing lymphedema after breast cancer depends on several factors, such as axillary node dissection, radiotherapy, or obesity, and it is also related to the anatomic variability between patients. ICG lymphography allows visualization of the anatomic patterns of the lymphatic system and their degeneration. It reveals how channels are more functional in some patients than in others. Patients with secondary drainage channels will be able to maintain better activity of the lymphatic system, and their risk of developing lymphedema may be lower. The differences among patients regarding these factors underscore the need to customize treatment to match the needs of individual patients.

Preoperative Assessment The key concept in decision-making regarding potential candidates for reconstructive surgical techniques for lymphedema is to identify patients who still have a remaining functional lymphatic system. Following an accurate clinical evaluation, assessment with diagnostic imaging techniques is essential to study the functional and morphologic features of the lymphatic system so that each patient can be offered the best therapeutic option.

Clinical Evaluation A full clinical examination is crucial in all potential candidates, in addition to a detailed medical history, so the lymphedema stage can be determined according to the International Society of Lymphedema.21 This evaluation allows differentiation between pitting and nonpitting edema. Nonpitting edema is characterized by severe fibrosis and hypertrophy of adipose tissue and indicates an advanced stage of the disorder. During the clinical examination, limb circumferences are measured at predetermined anatomic levels and photographic documentation is obtained.

K23349_Neligan_41_Combined Treat_r2_dc_0539-0552.indd 540

5/27/2015 12:48:06 PM

Chapter 41  Combined Surgical Treatment for Breast Cancer–Related Lymphedema

541

FIG. 41-1  ICG lymphography. ICG is captured by an infrared camera system, and functioning lymphatic channels can be visualized on a display device as fluorescent channels.

Diagnostic Imaging Techniques Diagnostic imagining techniques are needed to determine an appropriate therapeutic strategy for each patient. Our assessment protocol involves: indocyanine green (ICG) lymphography, lymphoscintigraphy, MR lymphography, and in some cases, CT angiography. For us, the essential technique is ICG lymphography, which we perform as a diagnostic test in the outpatient clinic during the first consultation. Depending on the results, we will plan to do additional imaging studies.

Indocyanine Green Lymphography We have found that ICG lymphography provides a decisive test image. This is the first preoperative test in our assessment protocol, since it shows the functionality of the lymphatic system. The procedure consists of injecting 0.1 to 0.2 ml of indocyanine green dye subcutaneously at the second and fourth interdigital web spaces of both arms. ICG is captured by an infrared camera system, and functioning lymphatic channels can be visualized on a display device as fluorescent channels (Fig. 41-1). ICG lymphography provides valuable information in our assessment, making it a double evaluation: • First, it allows the examiner to see immediately after injection the velocity of the contrast ascending to the axilla, and after 5 minutes where the contrast medium is stored, and the dermal backflow can be evaluated. Thus the exact location of the active lymphatic channels and transport capacity of them can be determined. With the use of ICG lymphography we have realized that there is a substantial anatomic and functional variability between patients, not only in the distribution and the number of lymphatic channels, but also in the velocity of transport of ICG.

K23349_Neligan_41_Combined Treat_r2_dc_0539-0552.indd 541

5/27/2015 12:48:07 PM

542

Part V  Current Treatment of Lymphedema

• Second, ICG lymphography provides valuable presurgical data about the degree of impairment in the lymphatic system. It identifies the number of enhanced lymphatic channels and their appearance and shows the lymphatic patterns and the presence and appearance of dermal backflow patterns. In addition, the uptake of ICG in a lymphatic channel and its distal to proximal transport means that the channel maintains its contractile function. This information is of crucial importance during the preoperative assessment, because only a patient with functioning lymphatic channels can be considered a potential candidate for LVA surgery. The preoperative planning assessment with ICG lymphography is done the day before LVA surgery or the same day in the operating room. It provides a map of the lymphatic system, and the lymphatic channels are marked on the patient’s skin surface. We complete the preoperative mapping with the information provided by MR lymphography, as explained later in more detail. ICG lymphography is also of paramount importance during ALNT surgery helping to understand the lymphatic pattern of the donor site limb. Because we normally use the superficial inguinal nodes as donor nodes for the transplantation in the axilla, during ALNT surgery lymphography allows us to localize the lymph nodes draining the inferior limb. This step reduces the risk to harvest a flap containing these lymph nodes, which would induce an iatrogenic lower limb lymphedema, as recently described.22 After LVA, ICG lymphography also helps us to assess whether the lymphatic transport function has improved compared with the preoperative examination, to evaluate the patency of an LVA, and to select new functional lymphatic channels when a secondary LVA surgery is planned. After ALNT, ICG lymphography can demonstrate the viability of autotransplanted lymph nodes when they are superficial (less than 2 cm deep). A considerable limitation of ICG lymphography is that it only allows exploration of the superficial tissue (1 cm), and when there is a diffuse pattern with dermal backflow in the first minutes of injection, a white image is seen that could encumber visualization if there are deeper channels, especially in obese patients or those with fat hypertrophy. As a result, we start to work in combination with MR lymphography.

Lymphoscintigraphy Lymphoscintigraphy is a standardized imaging test that provides good information on lymphatic function, as well as comparative information for postoperative assessment. Human serum albumin or sulfur colloid is injected into the first and second web space of each hand. The patient is asked to clench and release the fist repeatedly. Images are obtained with the patient in the supine position. Over the first 40 minutes and at 60, 120, and 180 minutes after the injection, dynamic images are taken of both arms, the axilla, and the liver. The main parameters evaluated are the tracer pathway, its appearance time at the axilla, the presence or absence of the major lymphatic collectors, visualization of nodes, and the presence or absence of dermal backflow. If axillary tracer uptake is absent, potential autologous lymph node transplantation to the axilla level could be planned. In a patient with upper limb lymphedema but who has remaining lymph node functionality at the axillary region, there is no reason for lymph node transfer in the axilla. The surgeon should consider some other technique or in some special cases might opt for autologous lymph node transplantation to the cubital fossa, or even to the wrist level.

K23349_Neligan_41_Combined Treat_r2_dc_0539-0552.indd 542

5/27/2015 12:48:07 PM

Chapter 41  Combined Surgical Treatment for Breast Cancer–Related Lymphedema

543

As an objective comparative follow-up method, further lymphoscintigraphy is performed 12 months after surgery to assess any improvement in drainage and the viability of the transplanted lymph nodes. Postoperative lymphoscintigraphy is also useful to verify the long-term patency of LVA. Indirect findings in respect to preoperative lymphoscintigraphy, such as a decrease in dermal backflow or the disappearance of the tracer in a site of an LVA from tracer passage into the blood circulation, can demonstrate patency of the LVA. Despite its advantages, lymphoscintigraphy does not provide detailed morphologic information about the lymphatic system, and findings must be complemented with ICG lymphography and MR lymphography.

MR Lymphography To gain even more information than that provided by ICG lymphography alone, in 2009 we began to include MR lymphography in our assessment. MR lymphography provides a three-dimensional reconstruction of the whole limb and shows both the superficial and the deep lymphatic system. It allows visualization of the lymphatic system when results of ICG lymphography are completely negative. We also perform MR lymphography once we have decided to proceed with surgery so that we obtain the best information and can plan the technique more efficiently and precisely. To map the lymphatic channels, hyperintense markers are placed on the skin surface at predetermined fixed points located along a reference line that goes from the acromion to the thumbnail, passing through the central point of the cubital fossa. Starting at the central point of the cubital fossa (point 0), markers are placed every 10 cm along the reference line. The data obtained not only provide a thorough assessment of a limb’s lymphatic system but also help to select the most suitable lymphatic channels for LVA. These should be as proximal as possible and have a good caliber and a beaded appearance. They can be distal in respect to a dermal backflow area, since this is considered a sign of lymphatic obstruction. Two coordinates for each lymphatic vessel are chosen and marked on the skin before LVA surgery. Combining the information provided by ICG lymphography and MR lymphography, the goal is to locate the contractile vessels most likely to promote successful LVA. Furthermore, with the use of MR lymphography it is possible to find on the deep level some lymphovenous shunts, which could otherwise lead to some confusion or misinterpretation. In other words, the superficial lymphatic channels are easily recognized.

Computed Tomography/Angiography We perform CT angiography only when ALNT is planned. It has become an essential tool in our practice to study the inferior epigastric donor area and to minimize the potential donor site morbidity in the lower limb. Based on our experience performing CT angiography, we have realized that there is significant variability in the number and location of superficial and deep inguinal nodes. We select the cranialmost and lateralmost inguinal nodes—first, because they are richly vascularized by the super-

K23349_Neligan_41_Combined Treat_r2_dc_0539-0552.indd 543

5/27/2015 12:48:07 PM

544

Part V  Current Treatment of Lymphedema

ficial epigastric system or the superficial circumflex iliac system, and second, to reduce the risk of secondary iatrogenic lymphedema. CT angiography helps to assess the number and location of superficial inguinal nodes to be harvested in the lymph node flap and evaluate the lymph node vascular pedicle (superficial inferior epigastric system or superficial iliac circumflex iliac vessels) to determine its size, course, and exact location by exact coordinates as described previously for abdominal perforator flaps.23

Surgical Techniques Lymphaticovenular Anastomosis LVA is generally performed under general anesthesia to avoid patient discomfort in view of the lengthy surgical procedure. The ICG lymphography and MR lymphography data guide the search for the most suitable, functioning lymphatic vessels. We start performing the LVA on the points of double coincidence, always considering first the proximalmost vessels. At the selected cutaneous point, we inject a small quantity of local anesthetic with epinephrine to reduce bleeding. We inject 0.1 to 0.2 ml of patent blue V about 2 cm distal to this point, because the dye often stains the lymphatic channels, making identification and dissection easier. A 2 to 3 cm cutaneous incision is then made and the lymph channels identified are carefully dissected and anastomosed end-toend or end-to-side to subdermal venules of similar caliber using 11-0 or 12-0 sutures, under a high magnification microscope (Fig. 41-2)

A

B

C

FIG. 41-2  A, Preoperative marking of the active lymphatic channels of the potential LVAs (green lines are the ICG information; red lines are per the MR lymphography). B, Terminoterminal anastomosis and terminolateral anastomosis between the distal stump of the lymphatic and the proximal stump of the venule. The lymph fluid can be seen flowing through the anastomosis along the vein. C, After the LVAs are completed.

K23349_Neligan_41_Combined Treat_r2_dc_0539-0552.indd 544

5/27/2015 12:48:07 PM

Chapter 41  Combined Surgical Treatment for Breast Cancer–Related Lymphedema

545

FIG. 41-3  Axial view of CT angiography showing the right superficial inguinal lymph nodes (yellow arrows) and the nourishing superficial circumflex iliac vessels (pink arrows). CT angiography facilitates location of the lymph nodes to be harvested and their nourishing vessels using a combination of coordinates in a system of cartesian axis.

In the immediate postoperative period (the first 48 hours), a gentle massage is performed every 2 hours on the area where the LVAs were done to promote drainage. Also it is strongly recommended that the affected extremity be elevated and the amount of activity limited. On postoperative day 3 we initiate muscular activation by having the patient perform isometric exercises with a rubber handball. At day 15, the patient may begin to swim or an AquaGym type of rehabilitation program three times per week for 1 year. We indicate manual lymphatic drainage for 1 year, twice a week during the first 6 months and once a week during the second 6 months.

Autologous Lymph Node Transfer The objective of using the ALNT technique is to replace the damaged or resected lymph nodes in the axilla with a vascularized free tissue transfer flap containing a few lymph nodes (between three and six nodes) from a nonrisk donor site. We prefer to use the superficial inferior epigastric area with superficial circumflex iliac vessels. Morbidity at this site is low, and the final cosmetic result is satisfactory because the scar is hidden (see Fig. 41-4). We study this area with CT angiography to assess the location of the superficial nodes and their vascular pedicle. We also determine the number and distribution of the deep lymph nodes to ensure that the ones we are removing will not disturb normal lymphatic lower limb drainage (Fig. 41-3). Before raising the flap (just before the skin incision), we inject 0.1 to 0.2 ml of ICG and 0.2 to 0.4 ml of 2.5% patent blue V dye intradermally in two or three spots above and below the inguinal fold in the potential drainage area to better visualize the superficial lymphatic vessels during dissection. To reduce the risk of seroma, we perform an accurate dissection, clipping and using bipolar cautery on the small lymphatic channels and side branches that go to the skin–adipose tissue–lymph node flap. With the lymph node flap we include a skin island of approximately 8 by 4 cm with some extra vascularized fat from the superficial circumflex iliac artery system. Its vascular pedicle is dissected gently up to the femoral vessels. The adipose tissue surrounding the lymph nodes and the skin island included in the flap are useful to replace the fibrotic tissue in the axillary region and also to facilitate the lymph absorption through the physiologic lymph-venous shunts.24,25 The skin island makes postoperative monitoring easier.

K23349_Neligan_41_Combined Treat_r2_dc_0539-0552.indd 545

5/27/2015 12:48:07 PM

546

Part V  Current Treatment of Lymphedema

Incision

Deep inferior epigastric and perforating (DIEP) vessels

DIEP flap

Lymphatic channels of the arm

Transferred lymph nodes

Internal mammary vessels

Vascularized lymph node transfer flap

DIEP vessels

Superficial circumflex illiac vessels

Thoracodorsal vessels

Femoral vessels Afferent vessels from transferred nodes Afferent vessels

Afferent vessels from upper limb

FIG. 41-4  The total breast anatomy restoration (T-BAR) concept involves immediate breast reconstruction with abdominal perforator flap and lymph-lymphatic anastomosis between the upper limb lymphatic vessels that have been cut during lymph node clearance and the afferent vessels from transplanted lymph nodes from the inguinal area.

Once the flap is isolated, the donor site is closed using a quilting suture to avoid dead spaces. We use a continuous spiral barbed suture, and we also spray the area with a tissue sealant to reduce the risk of seroma. A suction drain is left in the donor area until the drainage is less than 15 ml per day, and external compression is applied with use of a foam bandage for 2 weeks. Because the site we generally use for lymph node transfer is the axilla, anastomosis is normally performed between donor vessels of the circumflex scapular system after debridement of fibrotic tissue. It is essential to place the lymph nodes on the real apex of the axilla in contact with the axillary tissue; this is where the afferent dominant lymphatic channels arrive from the arm. If during dissection of the fibrotic tissue on axilla good lymphatic channels from the arm are found, a lymph-lymphatic anastomosis to the afferent lymphatic channels can be performed from the transplanted skin–adipose tissue–lymph node flap.

K23349_Neligan_41_Combined Treat_r2_dc_0539-0552.indd 546

5/27/2015 12:48:08 PM

Chapter 41  Combined Surgical Treatment for Breast Cancer–Related Lymphedema

547

To treat lymphedema and autologous breast reconstruction simultaneously with an abdominal free flap, we raise a compound abdominal (DIEP/SIEA) flap containing the lymph nodes with double vascularization (Fig. 41-4, A) and in some cases even with lymph-lymphatic anastomosis. This is the concept that we call the total breast anatomy restoration (T-BAR) technique (Fig. 41-4, B). On postoperative day 3 we initiate muscular activation by having the patient perform isometric exercises with a rubber handball. One month after surgery, the patient may begin to swim or an AquaGym type of rehabilitation program three times per week for 1 year. We indicate manual lymphatic drainage for 1 year, twice a week during the first 6 months and once a week during the second 6 months. The direction of the manual drainage should be toward the transplanted lymph nodes to stimulate neolymphogenesis. A light compression garment can be worn during the first 6 months; whether this will be needed for the entire time depends on the patient’s response.

Treatment Algorithm Appropriate patient selection is essential to successful BCRL treatment. Treatment must be individualized for every patient, regardless of the clinical stage or the evolution time of the lymphedema. A thorough preoperative study is necessary to determine the most potentially effective technique. Patients should be told that even in ideal cases the results may reduce the lymphedema by only a moderate amount. Patients may have high expectations for resolution of their condition, and the surgeon must be honest, explaining the limitations of the proposed surgical treatment. Decision-making is based on the results of the preoperative clinical assessment and imaging techniques (ICG lymphography, lymphoscintigraphy, and MR lymphography). Our current treatment algorithm is the combined lymphedema surgical treatment (CLST) (Fig. 41-5): • Nonfunctioning lymphatic channels (as determined by ICG assessment or MR lymphography) and nonpitting lymphedema: a reductive technique such as liposuction (the Brorson technique). Sometimes we can associate partial dermolipectomy simultaneously or in a second procedure. • Nonfunctioning lymphatic channels (as determined by ICG assessment or MR lymphography) and pitting lymphedema: intensive rehabilitation therapy; with reassessment of the possibility of a reductive surgical technique. • Functioning lymphatic system (as determined by ICG assessment and/or MR lymphography): we will choose reconstructive technique according to the clinical characteristics of the axilla: ȤȤ If the axillary area has a good status, we perform LVA in the affected limb. ȤȤ If the axillary area has abundant fibrotic tissue or signs of radiodermitis, we use an ALNT in the armpit after fibrotic tissue release, complemented by LVA in the affected limb distally. ȤȤ If the patient has amastia and requests breast reconstruction, we opt for ALNT with nodes included into the abdominal perforator flap (DIEP or SIEA), and we perform LVA distally on the affected limb.

K23349_Neligan_41_Combined Treat_r2_dc_0539-0552.indd 547

5/27/2015 12:48:08 PM

548

Part V  Current Treatment of Lymphedema

Preoperative assessment

Clinical examination

ICG lymphography

MR lymphography

Does lymphatic system still work?

No

Yes

Vibroliposuction

Simultaneous breast reconstruction required?

Yes

Abdominal PF 1 INT 1 LVA (T-BAR)

No No

LVAs

Yes

LNT 1 LVAs

Scarring tissue and/or radiodermitis in the axilla?

ICG lymphography Postoperative follow-up

Secondary LVAs Lymphoscintigraphy

FIG. 41-5  Treatment algorithm for combined lymphedema surgical treatment (CLST).

Further Considerations Investigators continue to search for an effective prophylactic treatment for BCRL; despite the reduction of incidence of lymphedema with sentinel node biopsy, obstructional lymphedema occurs in 5% to 9% of breast cancer patients.7 After studying more than 200 cases of patients with breast cancer–related lymphedema using ICG lymphography, we have observed that the distribution patterns of lymphatic channels vary

K23349_Neligan_41_Combined Treat_r2_dc_0539-0552.indd 548

5/27/2015 12:48:08 PM

Chapter 41  Combined Surgical Treatment for Breast Cancer–Related Lymphedema

549

among individuals. Patients with predominantly medial drainage without collateral dorsal vessels or with fewer than six lymphatic channels are more likely to develop lymphedema. In these patients, after mastectomy and axillary dissection, in an effort to prevent lymphedema we advocate the use of total restoration of breast anatomy (T-BAR) during the same operation. This involves immediate breast reconstruction together with lymph-lymphatic anastomosis between the upper limb lymphatic vessels that have been cut during node clearance and the afferent vessels from transplanted lymph nodes.

Conclusion Because the ideal surgical strategy for treatment of BCRL has yet to be established, the current key concept is to select patients for whom a surgical approach is likely to provide improvement and to choose the right technique for each patient. A detailed preoperative assessment should be performed to determine whether reconstructive surgery or palliative surgery is indicated. Only patients with minimal residual functionality of their lymphatic system will benefit from autologous lymph node transfer or LVA. Patients with irreversible hypertrophy of adipose and fibrotic tissue and absence of residual lymphatic activity should be offered a reductive technique. More research is needed on lymphatic anatomy, physiology, and physiopathology to gain better understanding of how lymphedema develops and to identify the best treatment in different stages. Investigators should make an effort to share their work and homogenize the preoperative assessment and treatment protocols to help to solve this terrible sequela of breast cancer treatment.

C linical P earls • Preoperative assessment is essential to correctly identify potential candidates for reconstructive surgical techniques. • ICG lymphography is the first preoperative test in our assessment protocol, since it shows the functionality of the lymphatic system. • MR lymphography provides three-dimensional reconstruction of the whole limb and shows both the superficial and deep lymphatic system. • We perform CT-angiography when axillary lymph node transfer is planned to minimize the potential donor site morbidity in the lower limb. • We select the most cranial and lateral inguinal nodes because they are richly vascularized by the superficial epigastric system or the superficial circumflex iliac system, thus potentially reducing the risk of secondary iatrogenic lymphedema.

K23349_Neligan_41_Combined Treat_r2_dc_0539-0552.indd 549

5/27/2015 12:48:08 PM

550

Part V  Current Treatment of Lymphedema

R EFERENCES 1. Hayes SC, Rye S, Battistutta D, et al. Upper-body morbidity following breast cancer treatment is common, may persist longer-term and adversely influences quality of life. Health Qual Life Outcomes 8:92-98, 2010. 2. Hayes S, Di Sipio T, Rye S, et al. Prevalence and prognostic significance of secondary lymphedema following breast cancer. Lymphat Res Biol 9:135-141, 2011. 3. Kwan W, Jackson J, Weir LM, et al. Chronic arm morbidity after curative breast cancer treatment: Prevalence and impact on quality of life. J Clin Oncol 20:4242-4248, 2002. 4. Paskett ED, Naughton MJ, McCoy TP, et al. The epidemiology of arm and hand swelling in premenopausal breast cancer survivors. Cancer Epidemiol Biomarkers Prev 16:775-782, 2007. 5. Ahmed RL, Prizment A, Lazovich D, et al. Lymphedema and quality of life in breast cancer survivors: The Iowa Women’s Health Study. J Clin Oncol 26:5689-5696, 2008. 6. Nesvold IL, Reinertsen KV, Fossa SD, et al. The relation between arm/shoulder problems and quality of life in breast cancer survivors: a cross-sectional and longitudinal study. J Cancer Surviv 5:62-72, 2011. 7. DiSipio T, Rye S, Newman B, et al. Incidence of unilateral arm lymphoedema after breast cancer: a systematic review and meta-analysis. Lancet Oncol 14:500-515, 2013. 8. Cormier JN, Askew RL, Mungovan KS, et al. Lymphedema beyond breast cancer: a systematic review and meta-analysis of cancer-related secondary lymphedema. Cancer 116:5138-5149, 2010. 9. Monleon S, Murta-Nascimento C, Bascuas I, et al. Lymphedema predictor factors after breast cancer surgery: a survival analysis. Lymphat Res Biol. 2014 May 16. [Epub ahead of print] 10. Charles RH. Elephantiasis Scroti. London: Churchill, 1912. 11. Becker C, Assouad J, Riquet M, et al. Postmastectomy lymphedema: long-term results following microsurgical lymph node transplantation. Ann Surg 243:313-315, 2006. 12. Koshima I, Inagawa K, Urushibara K, et al. Supermicrosurgical lymphaticovenular anastomosis for the treatment of lymphedema in the upper extremities. J Reconstr Microsurg 16:437-442, 2000. 13. Koshima I, Nanba Y, Tsutsi T, et al. Minimal invasive lymphaticovenular anastomosis under local anesthesia for leg lymphedema. Ann Plast Surg 53:261-266, 2004. 14. Campisi C, Eretta C, Pertile D, et al. Microsurgery for treatment of peripheral lymphedema: long-term outcome and future perspectives. Microsurgery 27:333-338, 2007. 15. Yamamoto T, Yamamoto N, Doi K, et al. Indocyanine green-enhanced lymphography for upper extremity lymphedema: a novel severity staging system using dermal backflow patterns. Plast Reconstr Surg 128:941-947, 2011. 16. Baumeister R, Siuda S. Treatment of lymphedemas by microsurgical lymphatic grafting: what is proved? Plast Reconstr Surg 85:64-74, 1990. 17. Rausky J, Robert N, Binder JP, et al. In search of an ideal surgical treatment for lymphedema. Report of the Second European Conference on Supermicrosurgery, Barcelona, March 2012. Ann Chir Plast Esthet 57:594-599, 2012. 18. Basta MN, Gao LL, Wu LC. Operative treatment of peripheral lymphedema: a systematic meta-analysis of the efficacy and safety of lymphovenous microsurgery and tissue transplantation. Plast Reconstr Surg 133:905-913, 2014. 19. Hadamitzky C, Pabst R, Gordon K, et al. Surgical procedures in lymphedema management. J Vasc Surg (in press). 20. Notohamiprodjo M, Baumeister RG, Jacobs TF, et al. MR lymphangiography at 3.0 T—a feasibility study. Eur Radiol 19:2771-2778, 2009. 21. Lohrmann C, Foeldi E, Langer M. MR imaging of the lymphatic system in patients with lipedema and lipo-lymphedema. Microvasc Res 77:335-339, 2009.

K23349_Neligan_41_Combined Treat_r2_dc_0539-0552.indd 550

5/27/2015 12:48:08 PM

Chapter 41  Combined Surgical Treatment for Breast Cancer–Related Lymphedema

551

22. International Society of Lymphology. The diagnosis and treatment of peripheral lymphedema: 2013 Consensus Document of the International Society of Lymphology. Lymphology 46:1-11, 2013. 23. Viitanen TP, Mäki MT, Seppänen MP, et al. Donor site lymphatic function after microvascular lymph node transfer. Plast Reconstr Surg 130:1246-1253, 2012. 24. Masia J, Clavero JA, Larrañaga JR, et al. Multidetector-row computed tomography in the planning of abdominal perforator flaps. J Plast Reconstr Aesthet Surg 59:594-599, 2006.

K23349_Neligan_41_Combined Treat_r2_dc_0539-0552.indd 551

5/27/2015 12:48:08 PM

K23349_Neligan_41_Combined Treat_r2_dc_0539-0552.indd 552

5/27/2015 12:48:08 PM

C hapter 42 Interventional Timing Peter C. Neligan, Jaume Masia

Inte

K ey P oints • Although there is no cure for lymphedema, early treatment improves outcomes. • Some patients respond to conservative treatment. • Patients should be referred early for surgery if they do not respond to conservative measures.

It has been made clear throughout this book that there is currently no cure for lymphedema. Although some treatments can yield spectacular results practically amounting to a cure, such results are inconsistent and exceptional. Evidence shows that early intervention leads to better outcomes.1 For this reason, it is important to develop a reasoned approach regarding which treatments to offer our patients, and when. The sooner a diagnosis can be made, it is more likely that we can develop a strategy and treatment plan before the condition becomes indolent and chronic. We also know that before lymphedema becomes clinically recognizable, there is a preclinical stage in which even the patient is unaware of the looming problem. Clearly, early detection is advantageous. Despite controversies in the literature, there is good evidence indicating that bioimpedance analysis (see Chapter 24) can detect lymphedema in this preclinical stage.2 It is possible that more widespread use of this technology as a screening tool may lead to earlier detection and consequently potentially better outcomes. Currently bioimpedance is considered experimental in many countries. The detection of preclinical lymphedema may also be possible with indocyanine green (ICG) lymphangiography.3 However, although it makes sense intuitively, much work must be done to confirm the link between early detection, early treatment, and better outcomes. This is particularly important in an era of constrained resources and greater accountability in health care. It is also essential to remember that some patients with postoperative lymphedema improve on their own, and invasive treatment should be avoided in this group of patients, although the use of conservative therapy at this stage reduces the risk of chronic lymphedema.4

553

K23346_Neligan_42_Interventional_r3_0553-0558.indd 553

5/26/2015 10:11:02 AM

554

Part V  Current Treatment of Lymphedema

However, should we look at our patients even before they undergo cancer surgery and try to determine which patients are most at risk of developing lymphedema? True, some individuals may develop lymphedema after sentinel node biopsy, whereas others do not, even after lymph node dissection and radiotherapy. There is some evidence in the literature that predicting which patients are at risk is not only possible but useful.5 One proposed approach to preventing the problem is to identify the draining lymph nodes of the arm or leg and preserve them during lymphadenectomy. This approach involves the concept of reverse lymph mapping and has been shown to be feasible in the upper6 and lower7 extremity. Some surgeons argue that we should treat patients prophylactically with lymphaticovenular anastomosis (LVA) at the time of lymph node dissection, and there is some proof to confirm that this may work.8 However, because the incidence of lymphedema after lymph node dissection is nowhere near 100%, such an approach means that we would be treating a large proportion of patients unnecessarily. Furthermore, the associated costs of such treatment (for example, resource use, intensity, and demand) make it untenable. It may be more reasonable to adopt such an approach in patients recognized as being at high risk of developing lymphedema. How do we identify this group of patients, and what treatment options do we offer them? The lymphatic microsurgical preventive healing approach (LYMPHA) procedure has been proposed as one such method in which the draining lymph nodes of the upper extremity are identified, and when removed, the afferent lymphatic channels to those nodes are anastomosed to a branch of the axillary vein (see Chapter 40).9 Although this is an attractive proposal, its utility is limited by the general lack of availability of such expertise and resources. Excluding prophylactic treatment, what other early treatment options can we offer our patients who have either preclinical or early stage clinical lymphedema? Conservative treatment is the first line of active therapy, and it is reasonable to suggest that every patient with early stage lymphedema should be offered a trial of conservative decongestant management. This is especially true, because lymphedema will resolve in a proportion of such patients. The big question is, when should we intervene with more radical treatment? Obviously, the patient who has tried and failed conservative treatment will be a potential candidate for surgical management, but how long do we wait to see if conservative treatment works? Furthermore, there is clear evidence that certain treatments are best managed at a particular stage of the disease. For example, liposuction (see Chapter 32) is indicated only after the chronic changes of fibrosis and increased fat deposition have become established. If the patient continues to have pitting edema (Fig. 42-1), indicating that there is a significant fluid element to the swelling, liposuction is not indicated. Conversely, we know that LVA is best performed before the stages of fibrosis and increased fat deposition, while there are still functioning lymphatic channels and a reasonable chance that LVA will work. We also know that the collecting lymphatic channels undergo a spectrum of changes ranging from normal through the ectatic, contracting, and sclerosing types and that these changes are indicative of the likely success of LVA.10 Akita et al11 conducted a prospective cohort observational study of 100 consecutive patients with gynecologic cancer who underwent pelvic lymph node dissection with ICG lymphangiography to assess early lymphatic flow patterns and risk factors. Of the patients who developed transient dermal backflow, those patients with a splash pattern of distribution tended to improve, whereas those with a stardust pattern did not. The authors concluded that perhaps these patients should be targeted for early treatment.11

K23346_Neligan_42_Interventional_r3_0553-0558.indd 554

5/26/2015 10:11:02 AM

Chapter 42  Interventional Timing

555

FIG. 42-1  Patient with lower limb lymphedema showing pitting edema that remains after the compressing thumb is removed.

For LVA to be successful, we must have functioning lymphatic channels. What do we offer the patient who does not have functioning lymphatic channels? Generally speaking, these are patients with established stage II or III lymphedema. Should we just offer them excisional procedures, including liposuction? We know that liposuction helps with limb volume reduction in these patients, even if they are still destined for lifelong compression.12 Vascularized lymph node transfer (VLNT) is another potential treatment. Can we reasonably expect transplanted lymph nodes to drain a limb in which there is no evidence of functioning lymphatic channels? There is controversy in the literature regarding where best to place the lymph nodes. Should they be placed distally where the edema is maximal13 or proximally, as proposed by Dr. Becker (see Chapter 36)? Cheng et al14 performed some elegant studies to show that lymph nodes act like sump pumps, and there is rapid transit of fluid from a flap containing lymph nodes into the draining vein of that flap. Regardless, VLNT is increasingly being used. However, a recent comprehensive review of the literature concluded that although the results with the use of VLNT for the treatment of lymphedema have largely been positive, further exploration into standardized protocols for the diagnosis, treatment optimization, and patient outcomes assessment is needed.15 With VLNT, there is much diversity in practice not just regarding placement of the lymph nodes but also whether to include a lymphaticolymphatic anastomosis, anastomosing an efferent lymphatic from the limb to an afferent lymphatic to the lymph node. Is that enough? How does the lymph drain from there? Does it drain to the bloodstream, or does it continue to drain to the main lymphatic pathways, and if so, how does it do that? Thus there are many unanswered questions, and our current recommendations may change as our knowledge increases. Our approach to patients with lymphedema is similar. Every patient who presents needs a diagnosis with many different methods of evaluation as already outlined in this book. It is also im-

K23346_Neligan_42_Interventional_r3_0553-0558.indd 555

5/26/2015 10:11:03 AM

556

Part V  Current Treatment of Lymphedema

A

B

FIG. 42-2  A, Patient with severe, long-standing lymphedema and a history of multiple previous episodes of cellulitis. B, Two years after festoon excision and multiple LVAs, she had a 40% reduction in limb circumference and no further episodes of cellulitis.

portant not to rely on any one modality. The patient in Fig. 42-2 has long-standing lymphedema with a history of many episodes of infection and trophic skin changes. Although it appears that she would not be a candidate for a bypass procedure, we found functioning lymphatics with ICG, performed several LVAs, and had a surprisingly positive outcome. Although we did not cure her lymphedema, she lost considerable limb volume, had a much lighter leg, significantly reduced her incidence of cellulitis, and overall was considerably improved. At the time we performed the LVA, she also had a direct excision of a festoon of lymphedematous tissue in her proximal thigh. We have already mentioned that patient history is of paramount importance. It is important to differentiate between primary and secondary lymphedema, postsurgical or postradiation lymphedema (or both), duration of the edema, previous treatments, and the patient’s response to previous treatments. This information can be gleaned from the history. The physical examination is also important and may guide the practitioner to the most appropriate treatment. In our practices we use imaging and consider it an important part of the evaluation, either with magnetic resonance lymphangiography (MRL) or ICG or both. The advantage of MRL is that it provides excellent information not only about the superficial lymphatics but also about the deep system. Furthermore, it helps to visualize where the edema is maximal and the state of fat deposition in the limb. Accurate volume measurements can also be gained from the MRL. The problem of identifying which is a lymphatic structure and which is a venous one has largely been eliminated.16 The downside of the technique is that it is time-consuming, technically demanding for the radiologist, and uncomfortable for the patient.

K23346_Neligan_42_Interventional_r3_0553-0558.indd 556

5/26/2015 10:11:04 AM

Chapter 42  Interventional Timing

557

Fluorescent lymphangiography with ICG is an extremely useful tool and is used extensively in lymphatic surgery. In fact, it is difficult to perform lymphatic surgery without ICG. In our practices it is used extensively both in diagnosis and treatment. The advantages of ICG are several. It is an easy technique and involves injection of ICG into the web spaces of the fingers or toes, excitation by a laser light source, and then examination with a near-infrared camera. When ICG is injected subcutaneously it is taken up by the lymphatics, making it a very useful tool for mapping subdermal lymphatic vessels. The one situation in which we proceed to schedule surgery is in patients with lymphedema who have a history of node dissection and radiotherapy. These are mostly postmastectomy patients. Some of these patients want to have their lymphedema treated and their breast reconstructed. In those patients we perform a deep inferior epigastric perforator flap in combination with lateral inguinal VLNT. Alternatively, in patients who do not want any reconstruction, we perform radical scar excision and VLNT, either from the inguinal region or from elsewhere, as described in Dr. Becker’s chapter on VLNT (see Chapter 36).

Conclusion Numerous options are available for both conservative treatment (see Chapter 29) and surgical treatment (see Chapters 30 through 37). There is good evidence that early intervention yields the best results. It is equally important to choose the correct modality of treatment. This choice is facilitated by a thorough history and preoperative imaging.

R EFERENCES 1. Boccardo FM, Ansaldi F, Bellini C, et al. Prospective evaluation of a prevention protocol for lymphedema following surgery for breast cancer. Lymphology 42:1-9, 2009. 2. Cornish BH, Chapman M, Hirst C, et al. Early diagnosis of lymphedema using multiple frequency bioimpedance. Lymphology 34:2-11, 2001. 3. Yamamoto T, Matsuda N, Doi K, et al. The earliest finding of indocyanine green lymphography in asymp­tomatic limbs of lower extremity lymphedema patients secondary to cancer treatment: the modified dermal backflow stage and concept of subclinical lymphedema. Plast Reconstr Surg 128:314e-321e, 2011. 4. Soran A, Ozmen T, McGuire KP, et al. The importance of detection of subclinical lymphedema for the prevention of breast cancer-related clinical lymphedema after axillary lymph node dissection; a prospective observational study. Lymphat Res Biol 12:289-294, 2014. 5. Diaconu C, Livadariu RM, Dogaru C. The risk of lymphedema after breast cancer surgical treatment. Rev Med Chir Soc Med Nat Iasi 116:1081-1086, 2012. 6. Klompenhouwer EG, Gobardhan PD, Beek MA, et al. The clinical relevance of axillary reverse mapping (ARM): study protocol for a randomized controlled trial. Trials 14:111, 2013. 7. Bats AS, Nos C, Bensaid C, et al. Lower-limb drainage mapping for lymphedema risk reduction after pelvic lymphadenectomy for endometrial cancer. Oncologist 18:174-179, 2013. 8. Boccardo F, De Cian F, Campisi CC, et al. Surgical prevention and treatment of lymphedema after lymph node dissection in patients with cutaneous melanoma. Lymphology 46:20-26, 2013.

K23346_Neligan_42_Interventional_r3_0553-0558.indd 557

5/26/2015 10:11:04 AM

558

Part V  Current Treatment of Lymphedema

9. Boccardo F, Casabona F, De Cian F, et al. Lymphedema microsurgical preventive healing approach: a new technique for primary prevention of arm lymphedema after mastectomy. Ann Surg Oncol 16:703708, 2009. 10. Mihara M, Hara H, Hayashi Y, et al. Pathological steps of cancer-related lymphedema: histological changes in the collecting lymphatic vessels after lymphadenectomy. PloS One 7:e41126, 2012. 11. Akita S, Mitsukawa N, Rikihisa N, et al. Early diagnosis and risk factors for lymphedema following lymph node dissection for gynecologic cancer. Plast Reconstr Surg 131:283-290, 2013. 12. Brorson H. From lymph to fat: liposuction as a treatment for complete reduction of lymphedema. Int J Low Extrem Wounds 11:10-19, 2012. 13. Lin CH, Ali R, Chen SC, et al. Vascularized groin lymph node transfer using the wrist as a recipient site for management of postmastectomy upper extremity lymphedema. Plast Reconstr Surg 123:12651275, 2009. 14. Cheng MH, Huang JJ, Wu CW, et al. The mechanism of vascularized lymph node transfer for lymphedema: natural lymphaticovenous drainage. Plast Reconstr Surg 133:192e-198e, 2014. 15. Raju A, Chang DW. Vascularized lymph node transfer for treatment of lymphedema: a comprehensive literature review. Ann Surg 261:1013-1023, 2015. 16. Mitsumori LM, McDonald ES, Wilson GJ, et al. MR lymphangiography: how I do it. J Magn Reson Imaging. 2015 April 23. [Epub ahead of print]

K23346_Neligan_42_Interventional_r3_0553-0558.indd 558

5/26/2015 10:11:04 AM

C hapter 43 Lymphedema Complications and Their Treatment Arin K. Greene, Reid A. Maclellan

K ey P oints • Besides swelling, lymphedema can cause numerous health problems. • Cardiovascular complications, although rare, can occur. • Genitourinary complications can accompany penoscrotal or vulval edema. Sexual function per se is not compromised, although it may be practically.

Lym

• Infection is one of the major and most common morbidities caused by lymphedema. • Malignant transformation is extremely rare but is associated with a very poor prognosis. • Massive localized lymphedema is not uncommonly associated with morbid obesity. In general, the obesity must be treated first. • Orthopedic issues can develop related to the weight of the lymphedematous tissue and the strain it puts on the skeletal framework. • Skin changes are very commonly associated with lymphedema and may precipitate soft tissue infections. • Psychosocial issues are very common.

Lymphedema is a chronic and incurable condition. Nevertheless, there are certain issues that we know and recognize about lymphedema. For example, patients who are compliant with interventions tend to have fewer problems than less compliant patients, although a few patients have severe morbidity.1 Active individuals have less complications than patients with a sedentary lifestyle. Muscle contraction aids the movement of lymph fluid through the body. Exercise slows disease progression by enhancing lymphatic flow and stabilizing the body mass index of patients with lymphedema. It is well documented that lymphatic function is negatively affected by obesity.2,3 We also know that individuals born with an anomalous lymphatic system have less difficulties from the disease than patients who develop lymphedema from an injury to their lymphatic vessels.1 Perhaps individuals with primary lymphedema counteract the debilitated lymphatic system 559

K23346_Neligan_43_Lymp Complications_r3_0559-0568.indd 559

5/26/2015 10:12:02 AM

560

Part V  Current Treatment of Lymphedema

BOX 43-1  Morbidity From Lymphedema • Cellulitis • Discomfort • Hyperkeratosis • Massive localized lymphedema • Lymphangiosarcoma • Lymphatic vesicles • Problems with the extremity • Progression of the disease • Psychosocial distress • Wearing clothing

better than patients with secondary disease. Morbidity from lymphedema is not insignificant (Box 43-1). The conditions discussed in this chapter are the most commonly seen complications from lymphedema.

Complications Affecting Specific Body Systems Complications of the Cardiovascular System Lymphedema does not affect the arteries or veins; therefore patients rarely develop congestive heart failure secondary to their disease. Nevertheless, there are exceptions (see Chapter 10).We have one patient with massive disease who had cardiac morbidity from lymphedema (Fig. 43-1). He presented with the most severe expression of primary lymphedema in our experience. The disease began in his right lower extremity, and over time the disease progressed to the point that he became nonambulatory. High-output congestive heart failure manifested as a result of excessive blood shunting to his right lower extremity and buttock. His congestive heart failure resolved after 45 pounds of skin and subcutis were excised from the diseased area.

Complications of the Genitourinary System Psychosocial morbidity is the main issue for patients with penile/scrotal lymphedema, because individuals feel that the affected area is unattractive. They may worry about their appearance in clothing or the difficulty of fitting into clothing because of the bulk of the disease. Vesicles can also form that leak and cause infection (Fig. 43-2). Dysuria or phimosis may occur; however, genital lymphedema does not influence sexual function or cause sterility per se. It is recommended that patients wear tight-fitting exercise shorts to increase compression to the diseased area. Leaking lymphatic vesicles can be sclerosed or treated with a carbon dioxide laser. Resection of the skin and subcutaneous tissue can alleviate the psychosocial morbidity by improving the appearance of the genitals.

K23346_Neligan_43_Lymp Complications_r3_0559-0568.indd 560

5/26/2015 10:12:02 AM

Chapter 43  Lymphedema Complications and Their Treatment

561

FIG. 43-1  This 32-year-old man had congestive heart failure resulting from primary lymphedema of his right lower extremity and buttock. There was massive shunting of blood through the diseased area.

FIG. 43-2  This 24-year-old man with primary lymphedema had low self-esteem because of the appearance of his genitals. He also had leaking lymphatic vesicles.

Complications of the Musculoskeletal System Lymphedema is primarily a disease of the skin and subcutaneous tissue. The bone is not directly affected; therefore children are not at risk for vertical limb overgrowth and do not need monitoring for a leg-length discrepancy. Patients may have a reduced ability to use an extremity because of muscle weakness and/or heaviness from overgrowth of the disease. Joint mobility may also be compromised by operative or radiation fibrosis. Compression therapy is prescribed for early lymphedema to decrease the size of the affected area and improve the ability to perform activities of daily living. As the disease progresses, lymph accumulation in the superficial interstitial space leads to subcutaneous adipose deposition.4 Patients initially increase muscle strength in the diseased extremity

K23346_Neligan_43_Lymp Complications_r3_0559-0568.indd 561

5/26/2015 10:12:02 AM

562

Part V  Current Treatment of Lymphedema

FIG. 43-3  Lymphedema can decrease the ability to perform activities of daily living. This 40-year-old woman with primary lymphedema has difficulty walking because of the weight of her left extremity.

FIG. 43-4  This 11-year-old boy with primary lymphedema of the lower extremity has vesicles of his feet, causing multiple infections.

because of the additional weight. Over time, the addition of fibroadipose deposition and muscle hypertrophy cause a decrease in function. Individuals may have difficulty dressing themselves, because they can no longer raise their arm over their head. The weight of lower extremity disease may impede ambulation (Fig. 43-3). Orthopedic morbidities are caused by severe disease exacerbated by fibroadipose deposition. Conservative interventions (for example, compression garments, pneumatic compression devices, and massage) do not address this problem. More aggressive treatments that decrease the weight of an extremity by removing excess skin and subcutaneous tissue help alleviate symptoms. Suction-assisted lipectomy or staged skin or subcutaneous tissue excision can reduce the excess weight of the extremity.

K23346_Neligan_43_Lymp Complications_r3_0559-0568.indd 562

5/26/2015 10:12:03 AM

Chapter 43  Lymphedema Complications and Their Treatment

563

Complications of the Integumentary System The appearance of the skin can be affected by lymphedema. Hyperkeratotic lesions or cutaneous lymphatic vesicles may form, causing psychosocial stress for some patients. Normally these skin changes affect the distal lower extremity1 (Fig. 43-4). Vesicles can also involve the scrotum or penis (see Fig. 43-2). Lymphatic vesicles may cause morbidity when they bleed or leak (lymphorrhea). As previously mentioned, lymphedema does not affect arteries or veins; therefore ulceration rarely occurs in patients with this disease. Patients may require treatment of their lymphatic vesicles because of psychosocial distress or continuous bouts of infection. The vesicles cause breaks in the skin and provide a route of entry for bacteria. Patients with multiple infections per year usually also have lymphatic vesicles. First-line therapy for the vesicles is sclerotherapy or a carbon dioxide laser. The injection of the vesicles with a sclerosant (for example, sodium tetradecyl sulfate) decreases leakage by causing fibrosis of the diseased area. A carbon dioxide laser also scars the vesicles. Resection usually is not performed because the vesicles often encompass a large area, and the distal extremity is an unfavorable location to excise skin.

Infection Infection is a common and potentially serious morbidity of lymphedema. The lymphatics function as an immunologic defense system; therefore a diseased area has impaired immunosurveillance. Lymph stasis creates a proteinaceous environment favorable for bacterial growth, which elevates the risk of developing cellulitis after minor trauma (Fig. 43-5). Functional lymphatics may be injured after an infection in a lymphedematous extremity, which can lead to further disease progression. In one community study, nearly one third of patients with lymphedema had an infection over the previous year and one fourth required hospitalization.5 In

FIG. 43-5  Patients with lymphedema are at increased risk for infection. This 18-year-old girl with primary lymphedema had an infection of the left lower extremity requiring hospital admission.

K23346_Neligan_43_Lymp Complications_r3_0559-0568.indd 563

5/26/2015 10:12:03 AM

564

Part V  Current Treatment of Lymphedema

another study of patients referred to a lymphedema program, 42% had a lifetime history of cellulitis, and 70% of those patients required in-patient intravenous antibiotics.6 An incidental break in the skin is the most common entry point of infection for patients with lymphedema. The patient normally is unaware of the cut or scrape. Infections in lymphedematous extremities can spread more quickly compared with areas without disease. Patients are counseled to wash and moisturize the extremity regularly to prevent desiccation and skin breakdown; protective clothing should be worn to avoid trauma. Sepsis may form swiftly from a superficial cellulitis; therefore patients are advised to seek immediate medical attention at the first sign of an infection. Patients who have three or more episodes of cellulitis per year may require chronic suppressive antibiotic therapy.

Malignant Transformation Stewart-Treves syndrome originally referred to lymphangiosarcoma that formed in breast cancer patients who had chronic lymphedema.7 Patients with chronic secondary lymphedema of the lower extremity, primary disease, or massive localized lymphedema also may develop lymphangiosarcoma7,8 (Fig. 43-6). The probability of an individual developing lymphangiosarcoma in an affected extremity because of chronic lymphedema is exceptionally low7 (about 0.07% to 0.45%). Mean survival is less than 2 years for patients with lymphangiosarcoma because of the high likelihood of metastases and local recurrence. Chemotherapy and radiation have minimal efficacy. Amputation of the diseased extremity can increase overall survival if metastases have not formed.7

FIG. 43-6  Lymphangiosarcoma in a lymphedematous arm. This 12-year-old boy with infant-onset primary lymphedema presented with a 3-month complaint of pain and hardening of his left hand and forearm. Biopsy results showed lymphangiosarcoma.

K23346_Neligan_43_Lymp Complications_r3_0559-0568.indd 564

5/26/2015 10:12:03 AM

Chapter 43  Lymphedema Complications and Their Treatment

565

FIG. 43-7  This 48-year-old man developed obesity-induced massive localized lymphedema of his scrotum.

Massive Localized Lymphedema Massive localized lymphedema is a rare condition that affects extremely obese patients. The disease is characterized by a large, localized area of overgrowth9 (Fig. 43-7). The mean patient body mass index is 61.8 Massive localized lymphedema mainly forms at the thighs and lower abdomen. Even when only one extremity has massive overgrowth, both legs may have malfunctioning lymphatics. Depending on the diseased area, patients might complain that tight-fitting clothing, ambulation, or even sitting is difficult. We refer all individuals to a bariatric center before surgical intervention is offered for their massive localized lymphedema. Operative morbidity and the recurrence rate of the diseased area are significantly higher if massive weight loss has not occurred first.8

Pain Lymphedema generally does not cause pain. Patients with severe disease of the lower extremity may feel pressure when wearing shoes. Excess weight can stress the joints of the diseased extremity. Patients may state that the limb feels heavy and causes fatigue or weakness. A patient most likely has another disease if a patient complains of significant pain.10

K23346_Neligan_43_Lymp Complications_r3_0559-0568.indd 565

5/26/2015 10:12:03 AM

566

Part V  Current Treatment of Lymphedema

Progression Lymphedema is a progressive disease. Stagnation of proteinaceous lymph fluid leads to inflammation that induces fibrosis and subcutaneous adipose tissue hypertrophy; over time the circumference of the limb can enlarge by 73%.4 The patient’s ability to use the area worsens as the diseased extremity increases in size.

Psychosocial Consequences of Lymphedema Many patients with lymphedema have difficulty fitting into clothes (Fig. 43-8). Individuals with only a single affected extremity have even more trouble because of the asymmetry. Feet and the lower extremity cause the greatest dilemma. Patients may have to wear two different shoe sizes to compensate, whereas others are forced to wear only open footwear (for example, sandals). Individuals with severe disease of the lower extremity find it difficult to fit into jeans or slacks and decide to wear loose-fitting pants. Patients with lymphedema of the upper extremity may have difficulty with fitting long sleeves; instead, they wear only short-sleeved or oversized shirts. Rings, watches, and bracelets may also need to be increased in size to fit appropriately.

FIG. 43-8  Fitting of clothing can be a problem for patients. This 55-year-old woman with secondary lymphedema of the lower extremity after an lymphadenectomy and radiotherapy for Hodgkin lymphoma has difficulty wearing pants because of the asymmetry of her legs.

K23346_Neligan_43_Lymp Complications_r3_0559-0568.indd 566

5/26/2015 10:12:04 AM

Chapter 43  Lymphedema Complications and Their Treatment

567

FIG. 43-9  Lymphedema can cause psychosocial morbidity. This 50-year-old man with adult-onset lymphedema of the left lower extremity has decreased self-esteem because of the appearance of his diseased limb.

Lymphedema can also negatively affect one’s psychosocial well-being. Individuals may feel that the disease makes them less attractive (Fig. 43-9). They might not like the appearance of their limbs or genitals, and this leads to a loss of self-esteem.11 The more severe the lymphedema, the greater the negative effects on self-esteem. Patients with unilateral involvement usually express even more dissatisfaction with their appearance because the asymmetry makes the lymphedema more noticeable. Individuals may attempt to keep their affected area covered at all times, which also negatively impacts their overall well-being.11 Children have difficulty comprehending their diagnosis and can feel different from their peers. Breast cancer survivors with lymphedema have a daily reminder of their malignancy. They score lower on quality-of-life measures compared with their counterparts who do not have lymphedema.12 For some people, the stigma of wearing compression garments is worse than the deformity. We advocate that children wear tight-fitting stockings that they choose instead of medical-grade compression garments. Counseling is offered to patients with psychosocial distress. Conservative compression treatments and surgical interventions can help the individual’s self-esteem by reducing the amount of asymmetry. Although it might appear that a swollen limb that is not painful does not create a major issue, nothing could be further from the truth. Lymphedema can have an impact on every aspect of one’s life, leading not only to major medical morbidities, but also to lifelong psychosocial issues that plague the individual for a lifetime.

K23346_Neligan_43_Lymp Complications_r3_0559-0568.indd 567

5/26/2015 10:12:04 AM

568

Part V  Current Treatment of Lymphedema

C linical P earls • Most patients with lymphedema do not suffer significant morbidity. • The most common problems caused by the disease are decreased self-esteem and infection. • Other sources of morbidity include lymphatic vessicles, lymphorrhea, musculoskeletal limitations, and malignant degeneration. • Patients are counseled to avoid incidental trauma to the diseased area and exercise. • Conservative management consists of compression regimens; a few individuals benefit from operative intervention.

R EFERENCES 1. Schook CC, Mulliken JB, Fishman SJ, et al. Primary lymphedema: clinical features and management in 138 pediatric patients. Plast Reconstr Surg 127:2419-2431, 2011. 2. Helyer LK, Varnic M, Le LW, et al. Obesity is a risk factor for developing postoperative lymphedema in breast cancer patients. Breast J 16:48-54, 2010. 3. Greene AK, Grant FD, Slavin SA. Lower extremity lymphedema and elevated body mass in obese patients. N Engl J Med 366:2136-2137, 2012. 4. Brorson H, Ohlin K, Olsson G, et al. Breast cancer-related chronic arm lymphedema is associated with excess adipose and muscle tissue. Lymphat Res Biol 7:3-10, 2009. 5. Moffatt CJ. Lymphoedema: an underestimated health problem. QJM 96:731-738, 2003. 6. Maclellan RA, Couto RA, Sullivan JE, et al. Management of primary and secondary lymphedema: analysis of 225 referrals to a center. Ann Plast Surg. 2014 Apr 8. [Epub ahead of print] 7. Sharma A, Schwartz RA. Stewart-Treves syndrome: pathogenesis and management. J Am Acad Dermatol 67:1342-1348, 2012. 8. Chopra K, Tadisina KK, Brewer M, et al. Massive localized lymphedema revisited: a quickly rising complication of the obesity epidemic. Ann Plast Surg. 2013 May 30. [Epub ahead of print] 9. Farshid G, Weiss SW. Massive localized lymphedema in the morbidly obese: a histologically distinct reactive lesion simulating liposarcoma. Am J Surg Pathol 10:1277-1283, 1998. 10. Brorson H, Ohlin K, Olsson G, et al. Quality of life following liposuction and conservative treatment of arm lymphedema. Lymphology 39:8-25, 2006. 11. Fu MR, Ridner SH, Hu SH, et al. Psychosocial impact of lymphedema: a systematic review of literature from 2004-2011. Psychooncology 22:1466-1484, 2013. 12. Swenson KK. Case-control study to evaluate predictors of lymphedema after breast cancer surgery. Oncol Nurs Forum 36:185-193, 2009.

K23346_Neligan_43_Lymp Complications_r3_0559-0568.indd 568

5/26/2015 10:12:04 AM

C hapter 44 Treatment of Lymphedema in India Rajiv Agarwal, Devisha Agarwal

K ey P oints • Lymphatic filariasis is the most prevalent form of lymphedema in India and is usually caused by Wuchereria bancrofti and Brugia malayi. • Medical treatment options for lymphatic filariasis include the administration of diethylcarbamazine in combination with albendazole. • Nonfilarial lymphedema typically presents as postmastectomy lymphedema, for which the mainstay of treatment is comprehensive decongestive therapy. • Surgical treatment comprising direct excision with skin grafting offers the best chance of improvement in patients whose lymphedema is refractory to conservative medical management.

Tre

Lymphedema is a permanent, progressive, and debilitating condition that develops from dysfunction of the lymphatic system. In India, filarial lymphedema is by far the most common form and is a public health problem for which an organized, national filarial control program is currently underway. Lymphatic filariasis affects 119 million people living in 73 countries; India accounts for 40% of the global prevalence of infection.1 Indigenous lymphatic filariasis has been reported mainly from Southern India, whereas the Northwestern states are free of indigenously acquired filarial infection. In India, lymphedema can be discussed in two broad categories: filarial and nonfilarial lymphedema. The justification for this categorization is the high incidence and prevalence of lymphatic filariasis in India to the point that it is endemic and necessitates independent consideration and importance. In mainland India, the causative nematodes are primarily Wuchereria bancrofti and Brugia malayi, which are transmitted by specific genera of mosquitoes that include Culex quinquefasciatus, Anopheles, and others. B. malayi infection is now reportedly restricted to rural areas of South India (Fig. 44-1). 569

K23346_Neligan_44_Treat in India_r2_dc_0569-0572.indd 569

5/26/2015 10:13:42 AM

570

Part V  Current Treatment of Lymphedema

FIG. 44-1  Prevalence of microfilariae associated with W. bancrofti in India. (Green districts, relatively lower incidence; yellow, 0.001% to 0.99%; red, 1% or above; white, no data.) There may be several pockets within a district with varying degrees of prevalence.

Treatment Options Past and Present The treatment options for lymphedema are broadly divided into two categories: medical and surgical. It is hard to distinguish between the past and present modalities of treatment because of the degree of intermingling in the various modalities. However, many past methods, such as crepe bandaging, and operations such as flap transfers, are now rarely used.

Nonsurgical Management Elimination of Lymphatic Filariasis Lymphatic filariasis is endemic in India. Consequently, a national filarial control program was launched in 1955 to control this disease.1 The objectives of the program were to delimit the problem, to undertake control measures in endemic areas, and to train personnel to staff the program. The main control measures were mass administration of an annual single dose of diethylcarbamazine, antilarval measures in urban settings, and indoor residual spray in rural areas. The strategies of this program included vector control, detection and treatment of filarial cases, and delimitation of endemic areas. In 1997 the World Health Organization and its member states made a commitment to eliminate lymphatic filariasis as a public health problem by 2020 through World Health Assembly Resolution WHA 50.29.1 The National Health Policy 2002, released by the Ministry of Health and Fam-

K23346_Neligan_44_Treat in India_r2_dc_0569-0572.indd 570

5/26/2015 10:13:42 AM

Chapter 44  Treatment of Lymphedema in India

571

ily Welfare, set the goal of the elimination of lymphatic filariasis in India by 2015. The twin pillars of lymphedema elimination strategy include transmission control and disability prevention and management. Mass drug administration with an annual single dose of 6 mg/kg body weight of diethylcarbamazine citrate (DEC) tablets in endemic areas resulted in a drastic reduction in the infection rate, microfilaria rate, and insignificant serious adverse effects in patients with lymphatic filariasis.3

Medical Treatment The specific drugs used for lymphatic filariasis are diethylcarbamazine in combination with albendazole. A single dose of 6 mg/kg of body weight of diethylcarbamazine is administered. Albendazole is also given as a single 400 mg dose, irrespective of age and weight. Diethylcarbamazine kills only about 50% of adult worms, and a single dose is as effective as a 12-day course against both the microfilariae and adult worms.4 Treatment with diethylcarbamazine also does not seem to reverse the lymphatic drainage after it is established.5 Diethylcarbamazine is a safe drug but may produce two types of side effects. The first is from the actual drug and may include headache, anorexia, nausea, abdominal pain, and vomiting; the second is from allergic reactions after the destruction of microfilariae and adult worms and may include fever, local inflammation, and pruritus.

Surgical Management Excision with skin grafting gives immediate and satisfactory long-term results in these patients (see Chapter 31). Excisional surgery has now become the treatment of choice for moderate-tolarge chronic filarial lymphedema of the leg and also for chronic genital lymphedema.6

Complications and Treatment Outcomes The most common complication of excisional surgery is graft loss and infection because of the high levels of bacteria residing on the skin of these patients. Minimal areas of graft loss are treated conservatively, and the wound is allowed to epithelialize. In patients with large, raw areas, repeat grafting may have to be undertaken to reduce morbidity.

Nonfilarial Lymphedema In India, nonfilarial lymphedema presents most commonly in the form of postmastectomy lymphedema.7 With the rapid increase in the early detection of breast cancer and with advancements in surgical techniques, surgical management of breast cancer has been increasingly adopted in India. This has resulted in an increase in the incidence of cases of postmastectomy lymphedema, which results from axillary dissection and radiation scarring of the axillary lymphatics in the treatment of breast cancer. This causes high morbidity with functional impairment, cosmetic disfigurement, and significant psychological effects.

K23346_Neligan_44_Treat in India_r2_dc_0569-0572.indd 571

5/26/2015 10:13:42 AM

572

Part V  Current Treatment of Lymphedema

The mainstay of treatment is comprehensive decongestive therapy (CDT) (see Chapter 29). CDT comprises an intensive phase of therapy from trained physical therapists, which includes manual lymphatic drainage, multilayered compression bandaging, exercises, and skin care. This ther­ apy leads to a significant reduction in the thickness of the skin and subcutaneous tissue that can be assessed with serial circumference measurements, water displacement volumetry, and highfrequency ultrasound. Therefore CDT is very effective in the long-term treatment of postmastectomy lymphedema, given the tropical climate, which may be a major factor in limiting the regular use of bandages by patients.

Conclusion Intensive, well-planned, and sequenced treatment of outpatients with all stages of lymphedema can produce a significant reduction in the volume of the edema over a short duration, even in the most advanced stages of the disease. Administration of antibiotics or diethylcarbamazine will not reduce the frequency of episodic adenolymphangitis. However, for the most part, simple hygienic measures, combined with good limb and foot care and local antibiotic or antifungal cream, if undertaken appropriately, are effective in reducing the number of adenolymphangitic attacks. Surgical treatment comprising direct excision with skin grafting offers the best hope for patients whose lymphedema is refractory to conservative medical management.

Acknowledgments The authors gratefully acknowledge the help of Professor Ramesh Chandra in proofreading the manuscript and Professor Padam K. Agarwal in helping with the histopathologic interpretation of the tissue specimens.

R EFERENCES 1. Ramaiah KD, Das PK, Michael E, et al. The economic burden of lymphatic filariasis in India. Parasitol Today 16:251-253, 2000. 2. Srivastava PK, Dhillon GP. Elimination of lymphatic filariasis in India—a successful endeavour. J Indian Med Assoc 106:673-674, 2008. 3. Vaishnav KG, Desai HS, Srivastava PK, et al. Impact of mass drug administration on elimination of lymphatic filariasis in Surat city, India. J Commun Dis 44:251-259, 2012. 4. Gyapong JO, Kumaraswami V, Biswas G, et al. Treatment strategies underpinning the global programme to eliminate lymphatic filariasis. Expert Opin Pharmacother 6:179-200, 2005. 5. Freedman DO, Bui T, De Almeida Filho PJ, et al. Lymphoscintigraphic assessment of the effect of diethylcarbamazine treatment on lymphatic damage in human bancroftian filariasis. Am J Trop Med Hyg 52:258-261, 1995. 6. Morey AF, Meng MV, McAninch JW. Skin graft reconstruction of chronic genital lymphedema. Urology 50:423-426, 1997. 7. Randheer S, Kadambari D, Srinivasan K, et al. Comprehensive decongestive therapy in postmastectomy lymphedema: an Indian perspective. Indian J Cancer 48:397-402, 2011.

K23346_Neligan_44_Treat in India_r2_dc_0569-0572.indd 572

5/26/2015 10:13:43 AM

C hapter 45 Surgical Treatment in China Ningfei Liu

K ey P oints • Treatment of lymphedema in China has traditionally been surgical. • Initially excisional operations were done. • Bypass procedures have been performed since the late 1970s. • Beginning in the early 2000s, liposuction, alone or in combination with microsurgery, became popular. • Today very few cases of microlymphatic surgery are performed. • New technologies, such as magnetic resonance lymphangiography, have enhanced surgeons’ ability to identify lymphatic structures and make specific diagnoses and help to guide appropriate treatment strategies.

Sur This chapter is a historical look at how Chinese surgeons have treated lymphedema over the past six decades. There is no detailed description of how these specific operations are performed. This review discusses both the successes and failures of various treatment approaches and examines the prospects for further improvements in the future. Lymphatic filariasis was the main cause of secondary lymphedema in China in the early 1950s. More than 20 million individuals were affected by filariasis, of which more than 5 million cases of lymphedema were caused by filariasis.1 The disease was predominantly found in agricultural villages, where most inhabitants were unable to receive timely treatment for the disease. In addition, inadequate treatment capabilities in these rural locations meant that by the time most patients, were diagnosed, they were already well into the advanced stages of the disease. At that time, the primary treatment method for lymphedema was surgical, and the Charles and Thompson procedures were commonly performed because elephantiasis was widespread. Because of the excision of large amounts of hyperplastic tissue, drastic reduction in bulk could be seen at the surgical sites, making the surgical results readily apparent. This is especially true for the Charles procedure, a method advocated by most surgeons at the time. In addition, to treat 573

K23349_Neligan_45_Surg Treat China_r4_0573-0580.indd 573

5/27/2015 1:18:30 PM

574

Part V  Current Treatment of Lymphedema

erysipelas, the village doctors in Fujian Province invented an “oven” made from bricks. The affected limbs were inserted into the oven and heated repeatedly, thereby significantly decreasing the rate of infection and to some extent the swelling. This method later developed into the socalled heating and bandage treatment.2 At the same time, there were scattered reports of only one or two documented cases of treatment involving transposition of the pedicle or free transplantation of the omentum to the axillary or inguinal region for draining lymphedema of the arm and leg.3 However, long-term follow-up showed no obvious effect on reducing lymphedema. During the 1970s, microscopic surgery for lymphedema treatment began to develop in China. In 1977 Dr. Di Sheng Chang4 was the first to present 11 cases of lymphaticovenous anastomosis. Of these 11, only 2 had good outcomes in the early stages. By early 1981, there were 60 cases reported from six surgical centers around the country. In 1988 more than 800 cases of lymphaticovenous anastomosis were reported worldwide. Among them, Chinese surgeons reported 300 such cases; lymphatic microsurgery had become very popular among the national surgeons. However, strict regulations were not enforced for case selection for lymphaticovenous anastomosis. Thus this surgery was performed for almost all stages of this disease, whether early, late, or primary or secondary. To create more anastomoses, three to four incisions were made in the affected limb, in which two to four lymphaticovenous anastomoses were applied at each surgical site. The reported clinical outcome of these microsurgical treatments on the lymphatic system was based solely on physical signs; the patency rate of lymphaticovenous shunting was not objectively proved in most reports. In the 1990s new and innovative procedures emerged for the surgical treatment of lymphedema, including the anastomosis of superficial and deep lymph vessels. Wei Gang Cao et al5 reported 8 successful cases of 13, whereas in the remaining 5 patients the surgeries were unsuccessful because the collecting lymph vessels (superficial or deep) were not found, which underscored the difficulties associated with the procedure. Several centers reported that anastomosis of the external iliac lymph vessels with the inferior epigastric vein was effective in alleviating chylous ascites.6 Based on a decade of clinical observations, surgeons reached an agreement: 1. Microlymphatic surgeries have a better clinical outcome for secondary lymphedema than for primary lymphedema. 2. Patients with secondary lymphedema for 5 years or less showed better treatment outcomes than patients who had the disease for more than 5 years. 3. Microlymphatic surgeries reduce the number of episodes of erysipelas. 4. For advanced cases of lymphedema, the Charles procedure was still preferred. After 2000, there was a decrease in the number of reported cases of lymphaticovenous microlymphatic surgeries. During this period, liposuction developed as a new treatment method for lymphedema of the extremity. It was used as a stand-alone operation for an entire limb or in combination with microsurgery.7 When combined with microsurgery, liposuction was performed at the distal part of the limb (lower leg or forearm), whereas free flap transplantation or lymphaticovenous anastomosis was done at the inguinal or axillary region.8

K23349_Neligan_45_Surg Treat China_r4_0573-0580.indd 574

5/27/2015 1:18:30 PM

575

Chapter 45  Surgical Treatment in China

To treat secondary arm lymphedema after radical mastectomy, flaps such as the latissimus dorsi and free abdominal flap transplantation to the axillary region were carried out at a few centers. Postoperative lymphoscintigraphic findings showed a decrease in dermal backflow in the affected limb.9 An alternative treatment method for breast cancer–related lymphedema of the arm was the transplantation of the great saphenous vein to bridge the deep and superficial lymphatic vessels.10 Five of nine patients showed an initial reduction of arm swelling. Xiaoxia Li et al11 were the first to describe the use of autotransplantation of bone marrow stem cells to treat lymphedema. Although the authors believed that bone marrow–derived stem cells have an effect on reducing lymphedema, there was no objective evidence (such as lymphatic system imaging) of regeneration or repair of lymphatic vessels in the treated limbs. An increasing number of patients who underwent the Charles procedure during the 1950s to 1990s returned to the hospital for treatment of their surgical complications, which included chronic ulcer, lymphorrhea, and disability of the ankle resulting from hypertrophic or atrophic scar. The lower legs that received split-skin grafts were often much smaller than the thighs. In addition to the affected external appearance, the surgical sites in some patients developed malignancies, such as squamous cell carcinoma or angiosarcoma. Unfortunately, the lack of viable alternatives meant that the Charles procedure was still the primary approach for many surgeons when patients reached the advanced stages. Since 2010, the number of reported microlymphatic surgeries has been dramatically reduced compared with the 1970s and 1980s (Fig. 45-1). At this writing, no more than three medical centers in China are routinely performing microlymphatic surgeries for the treatment of lymphedema. The Department of Plastic Surgery at the Shanghai Ninth People’s Hospital and the Beijing Plastic Surgery Hospital are attempting to treat arm lymphedema resulting from breast cancer treatments with vascularized lymph node transplantation or combining with free flaps to perform breast reconstructive surgery at the same time.12 Nevertheless, the number of reported cases still remains

400

380

300

Cases

231 200

100

62 12

0 1970

1980

1990

2000

Year

FIG. 45-1  Reported cases of microlymphaticovenous anastomosis during the 1970s to 2000s.

K23349_Neligan_45_Surg Treat China_r4_0573-0580.indd 575

5/27/2015 1:18:30 PM

576

Part V  Current Treatment of Lymphedema

in single digits. Furthermore, the long-term effects of the procedures are still under evaluation. The Department of Lymphatic Surgery at Beijing Shijitan Hospital has carried out thoracic duct surgery for more than 20 years to treat visceral lymphatic deformities, such as chylous reflux lymphatic malformations and chyloperitoneum. They have accumulated much experience in the field of thoracic duct surgery. Lymphaticovenous anastomoses combined with liposuction are also regularly performed to treat lymphedema of the extremity. In the history of surgical treatment of lymphedema in China, microlymphatic surgery was once the high-flying treatment in the 1970s to 1990s, followed by debulking procedures. Chinese surgeons may have performed the most of this type of procedure in the world. However, in recent years, progressively fewer surgeons have attempted these procedures, resulting in a net decrease in surgical case reports. The primary reason is low patient satisfaction with the outcome. The lack of knowledge regarding lymphatic disease and the limited capabilities of imaging technology meant that many microsurgical treatments for lymphedema were performed with a certain blindness. The surgeons may not have been completely clear about the morphologic and functional status of the lymphatic system in the lymphedematous limbs (for example, secondary lymphedema resulting from malignant tumor surgeries) before the lymphatic surgery. It is possible that the vessels were already partially obstructed or functionally impaired, resulting in an anastomosis that was bound to fail. Again, there was a lack of information on the type or subtype of primary lymphedema, such as pathologic changes in the lymph vessels or a combination of both lymphatic and lymph nodal disease. A surgical approach to reconstruct lymphatic pathways on a severely deformed system is impossible. The indications for microlymphatic surgery should not be based solely on the stage (early or late) of the disease. In recent years, medical imaging of the lymphatic system has advanced rapidly. The diagnostic value of new technologies, such as magnetic resonance lymphangiography (MRL), for evaluating the lymphatic system and the stage of disease has increased exponentially.13,14 High-resolution imaging and real-time observation of the structures and functional anomalies of the lymphatics and lymph nodes can help surgeons make a more precise diagnosis (Figs. 45-2 and 45-3) and accurately identify the type or subtypes of lymphatic pathologies (Fig. 45-4). Based on the more comprehensive evaluation of a patient’s lymphatic system that is made possible by MRL, appropriate surgery can then be selected, treatment can be much better individualized, and an improved curative effect can be expected. By the same token, the previous lack of high-resolution imaging also meant a lack of reliable tests to objectively evaluate the patency of lympha­ticovenous anastomoses or lymphatic regeneration after free lymph node transfers. Sometimes it was difficult to determine whether the decrease in patient bulk was from restoration of the lympha­tic circulation or from bandaging and bed rest in the hospital after surgery. The three-dimensional dynamic MRL may help provide an objective measure of clinical outcome and prognosis.

K23349_Neligan_45_Surg Treat China_r4_0573-0580.indd 576

5/27/2015 1:18:30 PM

577

Chapter 45  Surgical Treatment in China

D A

B

C

FIG. 45-2  Composition images of MR lymphangiograms show various lymphatic drainage pathways in primary lymphedematous limbs. A, Single deep lymph vessel (arrowhead) and popliteal nodes (arrow) were enhanced to show the absence of a superficial lymph vessel. B, Enhanced lymph vessels with cystic dilation (arrowheads) in the distal part of the leg. C, Both superficial lymphatics (arrowhead) and deep lymph vessels and popliteal nodes (arrow) were involved. D, A crisscross network of hyperplastic vessels in the thigh (arrow) and calf (arrowhead) are evident. A

B

C D

FIG. 45-3  Diverse inguinal node abnormalities of primary lymphedematous limbs visible on MR lymphangiograms. A, Enlarged inguinal lymph nodes (arrow) with homogeneous texture in the left side compared with lymph nodes of normal size in the right side. B, Single small node in a limb (arrow) with lymphatic hypoplasia and lymphedema is compared with lymph nodes in a limb without lymphedema. C, Partially contrast-enhanced inferior inguinal nodes (arrow) in bilateral lymphedema with secondary lymphatic dilation. D, Central part enhanced inferior inguinal node (arrow) compared with evenly enhanced contralateral healthy nodes.

K23349_Neligan_45_Surg Treat China_r4_0573-0580.indd 577

5/27/2015 1:18:30 PM

578

Part V  Current Treatment of Lymphedema

0 NORMAL

1 LYMPH NODE–AFFECTED ONLY Structural abnormalities

2 LYMPHATIC VESSELS AFFECTED ONLY

A

B

Aplasia or hypoplasia

Hyperplasia

3 LYMPH NODES AND LYMPHATIC VESSELS AFFECTED Aplasia or hypoplasia

A

B

Aplasia or hypoplasia

Structural abnormalities

Hyperplasia

C

Aplasia or hypoplasia

D

Aplasia or hypoplasia

Aplasia or hypoplasia

Hyperplasia

E

Hyperplasia

Structural abnormalities

F

Hyperplasia

Hyperplasia

FIG. 45-4  Updated classification of lymphatic system malformations in primary lymphedema. In lymph node–affected only classification, some moderate dilation of afferent vessels may also be present. 0, Normal. 1, Lymph node affected only. 2,  Lymphatic vessels affected only: A, Lymphatic aplasia/hypoplasia; B, lymphatic hyperplasia. 3, Both lymph node and lymphatic vessels affected: A, Lymphatic and nodal aplasia or hypoplasia; B, lymphatic aplasia or hypoplasia plus nodal hyperplasia; C, lymphatic aplasia/hypoplasia plus nodal structural abnormalities; D, lymphatic and nodal hyperplasia; E, lymphatic hyperplasia plus nodal aplasia or hypoplasia; F, lymphatic hyperplasia plus nodal structural abnormalities.

K23349_Neligan_45_Surg Treat China_r4_0573-0580.indd 578

5/27/2015 1:18:31 PM

Chapter 45  Surgical Treatment in China

579

The Present and the Future Currently surgery is no longer the major treatment for lymphedema. Instead, the trend has shifted toward more conservative methods, such as comprehensive decongestive therapy, to reduce edema and remodel the shape of an enlarged limb. Are surgeries no longer necessary for the treatment of lymphedema? The answer is certainly that surgery must remain an option. As long as the lymph nodes and lymphatic vessels are clearly identifiable and accessible, surgery should never be ruled out as a possible treatment choice. With further advancements in medical technology and as our knowledge of the lymphatic circulation system and pathologic mechanisms of lymphatic disorders increases, Chinese surgeons will strive toward the goal of discovering successful innovations in surgical procedures for the treatment of lymphedema.

C linical P earls • The pathology of the lymphatic system in primary lymphedema is complicated. The involvement of lymph vessels and/or lymph nodes should be considered when deciding between treatment options and should be dealt with accordingly. • Because of very bad outcomes, the so-called “debulking” procedure should not be the treatment of choice for lymphedema. • The surgeon should always consider the lymph vascular status before the start of surgical treatment for lymphedema, including secondary lymphedema.

R EFERENCES 1. Liu N, ed. Lymphedema—Diagnosis and Treatment. Beijing: Science Press, 2014. 2. Chang D. Heat and bandage treatment for chronic lymphedema of extremities; report of 1045 cases. Thai J Surg 3:3-12, 1985. 3. Meng X, Wu S. Two cases of pedicled omentum transplantation in the treatment of congenital scrotal lymphedema. Hebei Med J 6:322, 1984. 4. Chang D, ed. Plastic and Reconstructive Surgery. Shanghai: Shanghai Science and Technology Press, 1979. 5. Cao W, Huang W, Fu K. Anastomosis of superficial and deep lymphatics in the treatment of lower limb lymphedema. Chin J Reparative Reconstr Surg 8:137, 1994. 6. Geng W, Zhang L, Wu F. Microsurgical treatment of chylous ascites: report of four cases. Chin J Surg 33:462, 1995. 7. Cao W, Li S, Zhou SG. Liposuction in the treatment of primary lower limb lymphedema. Chin J Plast Surg 22:290-291, 2006.

K23349_Neligan_45_Surg Treat China_r4_0573-0580.indd 579

5/27/2015 1:18:31 PM

580

Part V  Current Treatment of Lymphedema

8. Yuedng S, Xuejun Z, Gu J. Flap transplantation combined with liposuction to treat upper limb lymphedema after mastectomy. Chin J Plast Surg 19:430-432, 2003. 9. Xiao N, Lu K, Shen S. Latissimus dorsi muscle flap transplantation in the treatment of breast cancer after radical operation of upper limb lymphedema. Chin J Plast Surg 16:7-9, 2000. 10. Li S, Chen S, Wang S. Treatment of arm lymphedema after mastectomy with vein graft. Shanghai Med J 23:393-395, 2000. 11. Li X, Yin Z, Jin X. Autologous bone marrow stem cell transplantation in the treatment of limb lymphedema. Chin Tissue Eng Res Clin Rehabil 12:6653-6656, 2008. 12. Mu L, Xin M, Liu C, et al. Vascularized lymph node transplantation for the treatment of breast cancer related lymphedema and breast reconstruction. Abstracts of the Eleventh National Conference of the Chinese Medical Association of Plastic Surgery Branch, Urumqi, China, Sept 2011. 13. Liu NF, Lu Q, Jiang ZH, et al. Anatomic and functional evaluation of lymphatics and lymph nodes in diagnosis of lymphatic circulation disorders with contrast magnetic resonance lymphangiography. J Vasc Surg 49:980-987, 2009. 14. Liu NF, Lu Q, Wu XF, et al. Comparison of radionuclide lymphoscintigraphy and dynamic magnetic resonance lymphangiography for investigating extremity lymphoedema. Br J Surg 97:359-365, 2010.

K23349_Neligan_45_Surg Treat China_r4_0573-0580.indd 580

5/27/2015 1:18:31 PM

Part VI

Research and Future Directions

K23349_Neligan_46_Animal Model_r4_dc_0581-0594.indd 581

5/27/2015 12:58:27 PM

K23349_Neligan_46_Animal Model_r4_dc_0581-0594.indd 582

5/27/2015 12:58:28 PM

C hapter 46 Animal Models of Lymphedema Swapna Ghanta, Daniel A. Cuzzone, Babak J. Mehrara

K ey P oints • Various animal models of lymphedema have been described. • Most animal models are secondary (injury-induced) lymphedema. • There is no perfect model, but available models can mimic the various stages of the disease process. • Large animal models are most useful for anatomic and histologic studies but are limited by costs and the availability of molecular reagents. • Small animal models are cost efficient and have widely available molecular reagents but are limited by the potential for surgical interventions.

An

Animal models of lymphedema have long been sought to increase our understanding of this disabling disease process. Historically, the goal of most models was to mimic secondary lymphedema, in which the lymphatic system was injured and lymphedema subsequently developed. However, recent advances in molecular biology have identified some genetic models of lymphedema that mimic primary (or congenital) forms of this disease.

Validity of Animal Models In general the currently available models of lymphedema are useful to study the pathologic process of this disease (Table 46-1). However, there are no perfect models that accurately match the clinical phenomena. All the available models have various deficiencies, including short-term and spontaneously resolving lymphedema, the need for extensive surgical manipulation, inherent animal morbidity, and additional manipulations such as radiotherapy or sclerosing agents to cause lymphatic injury, and the length of time required for the development of sustained lymphedema. These deficiencies occur because the lymphatic system has a significant built-in redundancy and excess capacity, the potential to regenerate and develop collateral vessels after injury, and the 583

K23349_Neligan_46_Animal Model_r4_dc_0581-0594.indd 583

5/27/2015 12:58:28 PM

584

Part VI  Research and Future Directions

TABLE 46-1  Animal Models Used for Study of Lymphedema and Lymphadenectomy Animal Model/Area of Investigation

Advantages

Disadvantages

Hindlimb

Relative size of lymphatic vessels and ease of cannulation and identification

Morbid surgical procedures; costly; lack of molecular reagents; lymphedema only in a subset of animals

Axilla

Size of lymphatics and radiologic analysis

Model of lymphadenectomy (not lymphedema)

Size of lymphatics and relative ease of surgery; lower expense than canine models; docile animals

Not available in many centers and limited or minimal swelling; more costly than murine models and lack of molecular reagents

Size of vessels; single inguinal node; easily identifiable afferent and efferent vessels

Relative costs (compared with murine models); relative lack of available molecular reagents

Docile animals; technically simple, nonmorbid procedure; reliable, sustained lymphedema; potential for therapeutic intervention

Relative costs (compared with murine models); nonanatomic location of lymphatic defects; relative lack of available molecular reagents

Hindlimb

Relatively inexpensive; some molecular reagents available; some transgenic strains available; reliable swelling (when surgery and radiation are combined)

Morbid procedures needed for lymphedema development and late skin fibrosis and breakdown

Tail

Simple procedure; radiographic evidence of disturbed lymphatic flow; relatively inexpensive; molecular reagents and transgenics available

Minimal swelling resulting from low skin compliance

Axillary dissection

Same as above

Minimal swelling at rest; requires exposure to irritants to elicit swelling

Tail

Simple procedure; well tolerated; reliable lymphedema lasting as long as 12 weeks; histologic findings similar to clinical lymphedema; transgenics and molecular reagents widely available; lowest animal and husbandry costs

Small lymphatic vessels complicate cannulation or isolation; nonanatomic location complicates use for surgical interventions; conditions that worsen wound healing secondarily worsen lymphedema

Hindlimb

Low cost

Morbid; lymphedema is not sustained

Axillary dissection

Simple; well tolerated; low costs; transgenics and molecular reagents available

No swelling or obvious histologic changes

Canine

Ovine: Sheep Lymphadenectomy

Porcine: Pig Lymphadenectomy

Lapine: Rabbit Ear

Murine: Rat

Murine: Mouse

K23349_Neligan_46_Animal Model_r4_dc_0581-0594.indd 584

5/27/2015 12:58:28 PM

Chapter 46  Animal Models of Lymphedema

585

development of spontaneous lymphatic bypass channels (for example, spontaneous lymphaticovenous anastomoses) after injury. Understanding the natural history of clinical lymphedema also illuminates why it is difficult to develop animal models of lymphedema. In developed countries, secondary lymphedema occurs most commonly after lymph node dissection for cancer treatment. In these procedures the lymph nodes and deep collecting system of the limb are damaged; however, the remaining soft tissues and superficial lymphatic network remain intact. As a result, the mild edema that develops in most patients immediately after surgery almost always resolves in a few weeks, and the affected limb is clinically indistinguishable from the normal limb. In a subset of patients, approximately 10% to 30%, chronic lymphedema develops months (and sometimes years) later either spontaneously or precipitated by a secondary insult, such as radiotherapy, infection, or trauma.1 In these patients, lymphedema is typically progressive and requires continuous management. The delayed onset of lymphedema (in a subset of patients) is critical to the inherent difficulties in the development of animal models, because the vast majority of researchers have tried to develop models in which chronic lymphedema forms immediately after surgery in 100% of animals. This goal does not match the clinical natural history of lymphedema (that is, delayed development in a subset of patients) and as a result has complicated the development of suitable models. Many of these models necessitate extensive surgery or the addition of other interventions, thereby increasing their complexity and morbidity. Nevertheless, various models have been described, which have been useful to study the pathologic findings of lymphedema. As such, the goal of researchers who study the pathologic processes of lymphedema is to select an animal model that balances animal morbidity, husbandry costs, and the availability of molecular reagents.

Surgical Models of Lymphedema Several reports have detailed the surgical techniques required to develop various animal models of surgically induced lymphedema in a large number of species, including dogs,2-6 sheep,7 rabbits,8 rats,9-11 and mice.11-13 In general, these models involved injury to the lymphatic system (usually both the superficial and deep lymphatics) and can be used for the histologic and radiologic study of the pathologic process of lymphedema (larger animal models) or with molecular techniques (small animal models).

Canine Limb Models The use of dogs to develop models of chronic lymphedema has a long history and dates back to Halsted,14 who was interested in this disease after he popularized the radical mastectomy. However, despite multiple attempts, Halsted was unable to develop a chronic model of lymphedema that was sustained over the long-term despite extreme surgical resections. Reichert15 and Drinker et al2 expanded on these efforts and removed the lymph nodes and deep soft tissues. Drinker et al repeatedly cannulated the deep lymphatics with sclerosing agents. Eventually, chronic lymphedema developed in a subset of animals at the cost of severe surgical morbidity and mortality and in most cases, multiple surgical interventions. Olszewski et al3 understood the importance of the superficial lymphatic network and described a canine model in which the deep lymphatics

K23349_Neligan_46_Animal Model_r4_dc_0581-0594.indd 585

5/27/2015 12:58:28 PM

586

Part VI  Research and Future Directions

were ligated at the midthigh, and a circumferential strip of skin, subcutaneous tissues, fascia, and periosteum was excised (to remove the superficial lymphatics), leaving a gap for healing by secondary intention. They followed 23 dogs over 1 year and found that all animals had initial edema that resolved in 6 to 8 weeks. However, in 8 of 23 animals (that is, 35%), chronic lymphedema developed 7 to 10 months later, which was confirmed by limb volume measurements, histologic findings, and radiologic analysis. Danese et al4 reported a canine hindlimb model that combined surgery and radiation treatment. In this model, the animals were initially treated with 1500 rad from the midthigh to the right lower quadrant, followed by surgical excision 2 to 4 weeks later. During surgery, the skin flaps were elevated from the abdomen to knee and muscle connective tissues, and the periosteum was removed, leaving only the skin bridge and neurovascular bundle intact. The deep lymphatics were visualized and removed for a length of at least 5 cm. These animals had initial edema that resolved, and approximately 50% developed chronic lymphedema at a later stage. Chen et al5 also described a radiation and surgical model in which the animals for the most part developed modest lymphedema (15% increase in volume). However, the combination of surgery and radiation was morbid, with postoperative infection and mortality rates of about 30%. Suami et al6 reported on lymph node dissection of a canine forelimb model. In this model, which used detailed radiologic methods and indocyanine green (ICG) lymphangiography, they described the lymphatic anatomy of normal dogs and also the anatomy 6 months after axillary lymph node dissection. Although axillary lymphadenectomy in this model did not result in long-term differences in limb volume or overt lymphedema, they noted a rerouting of the lymphatic system in the axilla, with bypass of the damaged lymphatic vessels through the superficial network. In general, the main reason for the use of dog models is the relative size of the lymphatic vessels and supporting structures, enabling the identification, cannulation, and radiologic analysis of the lymphatic tree with or without injury. The limitations of the described dog models include the morbid procedures necessary to produce lymphedema, the time required for lymphedema to develop, the development of lymphedema in only a subset of animals, the high costs of canine experiments (for example, animal husbandry and surgical procedures), and the lack of molecular reagents for canines.

Ovine Lymphadenectomy Models Tobbia et al16 reported a sheep model of lymphadenectomy in which the popliteal lymph nodes were removed and the afferent and efferent lymphatics were surgically ligated. These animals developed mild hindlimb edema (20% to 30% increase) that declined spontaneously over time, but in most animals remained higher than control limb measurements, even 16 weeks after surgery. Two months after lymphadenectomy, the animals had decreased lymphatic uptake of iodine-125. However, this value was normal (suggesting that the lymphatics had regenerated) by 4 months. Similar to the observations reported by Suami et al, Tobbia et al found histologic and immunohistochemical evidence of newly formed lymphatic vessels that bridged the gap left by lymphadenectomy. In other studies with this model, these authors17 used vascular endothelial growth factor C (VEGF-C) and angiopoetin-2 gene therapy in combination with lymph node transfer to study the effects of lymphangiogenesis in lymphatic functional recovery. In general, the advantage of the sheep model over the canine model is the relatively lower expense. However, the sheep

K23349_Neligan_46_Animal Model_r4_dc_0581-0594.indd 586

5/27/2015 12:58:28 PM

Chapter 46  Animal Models of Lymphedema

587

model has several disadvantages, including the limited availability of animals to various laboratories, limited limb swelling, no previous analysis of histologic changes, and the lack of available molecular reagents.

Porcine Lymphadenectomy Models In nearly identical articles, Lahteenvuo et al18 and later Honkonen et al19 described a porcine model of lymph node transfer in which the inguinal lymph node was identified and the inguinal lymphatic network (both proximally and distally) was ablated over 3 cm, leaving the vascular pedicle intact. The lymph nodes were not removed but left in situ and were vascularized to mimic microsurgical lymph node transplantation. The nodes were then injected with a control virus or an adenovirus expressing VEGF-C or VEGF-D. The authors did not report changes in limb volume (because none would be expected). However, they noted increased lymphangiogenesis and ingrowth of functional lymphatic vessels in animals treated with VEGF-C or VEGF-D. They described this procedure as a model of lymphedema; however, because there were no substantial, sustained changes in limb volumes or histologic measures of lymphedema and the lymph node was not removed, this is more accurately described as a model of lymphatic injury. Blum et al20 reported on a lymphadenectomy model in mini-pigs in which the superficial lymph nodes were excised. In some animals, the wound was simply closed; in others, the lymph nodes were transplanted as nonvascularized grafts in subcutaneous or submuscular pockets. As expected, there were no changes in limb volume, and all wounds healed satisfactorily. In most animals, technetium-99 (99Tc) drainage was also mostly normal, and only a few animals developed dermal backflow. Blum et al reported some integration of the transplanted lymph node and qualitative improvements in lymphatic transport capacity. However, these changes were not quantified. Based on the scant studies available in the pig model, the primary advantage of this model is the relative ease in identifying the inguinal lymph node and afferent and efferent lymphatics. The disadvantage is the relative cost of the animals, animal husbandry, and the lack of available molecular reagents.

Rabbit Ear Model The rabbit ear has been used in numerous lymphedema studies. This model with Japanese white rabbits was first reported in 1983 by Gong-Kang and Yuan-Pai.8 In this model, a 2 cm circumferential strip of skin, subcutaneous tissues, and perichondrium was resected at the base of the ear. In addition, the lymphatic collecting vessels (five to seven) were identified and resected. The neurovascular bundle and cartilage were preserved, and the wound was allowed to heal secondarily. The procedure was well tolerated by the animals, with only a few cases of necrosis resulting from injury to the neurovascular bundle. The animals developed significant edema of the ear (peaking at an increase in thickness of 400% and a volume increase of 330% by postoperative day 7), and the ears remained swollen for as long as 180 days. On histologic examination, the skin sections demonstrated inflammation (which subsided by postoperative day 20), increased numbers of fibroblasts and tissue fibrosis, hyperkeratosis, dense fibrosis of the stratum corneum, and obliteration of small lymphatics. This model has since been used in various therapeutic studies, including a study of lymph node transplantation,21 the delivery of recombinant human VEGF-C,22 and extracorporeal shock wave therapy.23

K23349_Neligan_46_Animal Model_r4_dc_0581-0594.indd 587

5/27/2015 12:58:28 PM

588

Part VI  Research and Future Directions

The advantages of the rabbit ear model include the use of a docile, relatively small animal, technical simplicity and reproducibility, sustained lymphedema, histologic changes similar to clinical secondary lymphedema, and the potential for therapeutic intervention. The disadvantages include relative costs (compared with rodents), nonanatomic location of the lymphatic defect (for example, lack of lymph node resection in the model), and relative lack of molecular reagents.

Rat Models Hindlimb Wang and Zhong24 applied the method of Olszewski et al to the rat hindlimb. In this model, a 1 cm circumferential skin excision, including the subcutaneous tissues. was performed at the midthigh. The main lymphatic trunks were identified and resected for a length of 1.5 cm, and the popliteal lymph node, together with its afferent and efferent lymph nodes, were removed. The skin was then sutured to the muscle, and the gap was allowed to heal secondarily. The authors had a fairly high operative mortality rate (21%), but reported that leg edema peaked by day 6 and then subsided to normal levels by day 30. However, by day 180 the animals developed mild (increase of approximately 8%) but statistically significant swelling of the lower extremity that persisted until day 270. The animals also had dilated lymphatic vessels distal to the zone of occlusion, dilated capillary lymphatics, fibrosis (in some samples), and skin thickening. Kanter et al9 modified the model proposed by Wang and Zhong in Sprague-Dawley rats by pretreating the animals with a single dose of 4500 rad of ionizing radiation 1 to 2 weeks before surgical lymphatic excision. Kanter et al also reported significant leg swelling, which was similar to the findings of Wang and Zhong. However, their final day of analysis was postoperative day 20, which made it difficult to determine whether the swelling persisted chronically. In addition, it was unclear whether the radiation increased the morbidity of the procedure. Lee-Donaldson et al25 reported a similar model in which radiation and surgery were combined. They found that the combination of these two insults, regardless of their timing, resulted in moderate to severe chronic lymphedema. However, these treatments were morbid and were associated with a 9% to 13% early mortality rate and late skin breakdown. Anthony et al10 described a microvascular hindlimb transplantation model in a rat. In this model, a unilateral hindlimb was autotransplanted and analyzed with 99Tc lymphoscintigraphy. These authors reported that swelling resolved quickly and 99Tc clearance was normal by day 12. However, this pattern appeared to occur as a result of multiple, small, superficial channels that bypassed the deep lymphatics. This pattern, which was similar to that described by Suami et al,6 persisted for a prolonged period (160 days).

Tail Slavin et al11 reported on a rat tail (and also a mouse tail) model of lymphedema that was used to study flap transfers. In this model, the lymphatic vessels at the base of the tail were identified and excised through a circumferential skin incision. The edges of the skin incision were cauterized, resulting in a 2 to 4 mm gap that was allowed to heal through secondary intention. It was difficult for the authors to assess lymphedema in the rat tail because of its low compliance and resultant limited swelling. However, lymphoscintigraphic analysis demonstrated minimal uptake

K23349_Neligan_46_Animal Model_r4_dc_0581-0594.indd 588

5/27/2015 12:58:28 PM

Chapter 46  Animal Models of Lymphedema

589

in the sacral nodes with dermal backflow. Technetium uptake was markedly improved with flap transfer to “bypass” the region of lymphatic obstruction. Because of the limited edema in the rat tail, this model is infrequently used, although Saito et al26 used it to show that transfection with hepatocyte growth factor improved secondary lymphedema.

Axillary Dissection Mendez et al27 reported on a rat axillary dissection model of lymphatic insufficiency. In this model, the axillary lymph nodes were identified and resected to mimic the clinical scenario. Seventy-three days after surgery, operated animals had minimal baseline limb swelling. However, when these animals were exposed to a skin irritant (oxazolone), they had more severe inflammation-induced edema and diminished lymphatic clearance (as assessed by ICG lymphography and quantification of interstitial transport) compared with sham operated animals.

Mouse Models Tail The mouse tail model of lymphedema has been reviewed extensively in the scientific literature and was first reported by Slavin et al.11 This model was almost identical in design to the rat tail model but was reported to be easier for volume changes because of the higher compliance of the mouse tail (and resultant increased swelling). The authors reported diameter increases of approximately 20% that persisted for as long as 100 days in nude mice. Rutkowski et al12 modified this model with the use of immunocompetent mice, and in a series of experiments they showed that lymphatic regeneration across the wound is regulated by the gradients of VEGF-C expression that guide and modulate the migration of lymphatic endothelial cells. In addition, these authors showed that the model, which was similar to clinical lymphedema, resulted in adipose deposition, lymphatic dilation, and chronic inflammation. The tail model was further modified by Clavin et al,13 who excised a 4 mm circumferential area of skin in the midportion (rather than at the base) of the tail to remove the superficial lymphatics and microsurgically ligate the deep lymphatics along the length of the wound. This modification enabled an analysis of gradients of tissue changes proximal and distal to the zone of injury. With this model, Avraham et al28 and Zampell et al29showed through a series of publications that the distal tail region remained chronically swollen for extended periods (as long as 12 weeks), and the histologic changes that occurred subsequent to injury were similar to the clinical scenario, including inflammation, fat deposition, fibrosis, and hyperkeratosis. This model is particularly useful because mice are relatively inexpensive, the surgical procedure is straightforward, lymphedema is reproducibly and chronically produced, and there is a wide availability of molecular reagents and transgenic mice available for analysis. However, the mouse tail model has limited use in studies designed to analyze lymph node transfers and depends on wound healing at the excision site. Therefore conditions that worsen wound healing could secondarily worsen lymphedema.

Hindlimb Chung et al30 described a mouse hindlimb model that was similar in design to the rat hindlimb. In this model, methylene blue was injected in the distal foot to identify the lymphatics. The skin,

K23349_Neligan_46_Animal Model_r4_dc_0581-0594.indd 589

5/27/2015 12:58:28 PM

590

Part VI  Research and Future Directions

subcutaneous tissues, deep lymphatics, inguinal lymph nodes, and hindlimb muscles were then resected with electrocautery. The neurovascular bundle was kept intact. The animals developed significant swelling as assessed by volume displacement shortly after surgery (postoperative day 1). However, this swelling decreased substantially by the first postoperative week and resolved completely by 1 month. In another hindlimb model described by Oashi et al,31 mice were exposed to 3000 rad in the left groin. One week later, a circumferential skin incision was made in the inguinal region. The deep lymphatic vessels were ligated, the subiliac and popliteal lymph nodes were removed, and the skin was sutured to the underlying skin, leaving a 1 to 2 mm circumferential gap. This led to mild lymphedema, with a limb circumferential length increase of 110% to 135%, which lasted up to 6 months. In lymphedematous limbs, ICG imaging showed a lack of clear lymphatic vessels and dermal backflow, which persisted for 6 months compared with the normal hindlimb, which showed flow to the popliteal lymph node.

Axillary Dissection Zampell et al32 described a mouse axillary dissection model in which the axillary lymph nodes were identified with methylene blue and excised along with the surrounding fat pad. This treatment resulted in mild arm edema (5% increase in volume) that persisted for 3 weeks but resolved thereafter, even though the lymphatic transport capacity of the limb was abnormal and dependent on collateral circulation. The authors also used this model to show, similar to clinical lymphedema samples and the tail model of lymphedema, that even 6 weeks after lymphadenectomy, the subcutaneous tissues of the distal extremity remained chronically inflamed and infiltrated with CD41 cells. Thus, although the axillary lymph node dissection model did not result in persistent swelling or fibrosis, the model had long-term impairment of the lymphatic transport capacity and was useful for corroboration of the findings of the mouse tail model.29 In addition, the axillary dissection model has been used successfully to study the molecular mechanisms of lymph node transplantation and lymphangiogenesis.

Genetic Models of Lymphedema Several genetic models of lymphedema have been described. These mice were discovered based on improvements in our understanding of the physiology and embryology of the lymphatic system. In general, these mice are models of primary lymphedema and have been used to study the immunologic or metabolic changes that occur from the abnormal development of the lymphatic system. A few studies have also evaluated the potential for therapeutic interventions in these mice as a means of modeling treatment approaches for primary lymphedema. The earliest description of a genetic model of lymphedema was a mouse with a spontaneous inactivating mutation of the vascular endothelial growth factor receptor 3 (VEGFR-3) gene. VEGFR-3 is a tyrosine kinase receptor that binds VEGF-C and VEGF-D and is known to play a prominent role in the regulation of lymphangiogenesis and lymphatic development. Mice deficient in both copies of VEGFR-3 die in utero because of severe abnormalities and lack of development of the lymphatic system. Humans with heterozygous mutations of VEGFR-3 develop primary lymphedema (Milroy disease), with hypoplastic cutaneous lymphatic vessels and impaired lymphatic

K23349_Neligan_46_Animal Model_r4_dc_0581-0594.indd 590

5/27/2015 12:58:28 PM

Chapter 46  Animal Models of Lymphedema

591

clearance. Karkkainen et al33 described a Chy-mutant mouse (presumably named because they develop chylous ascites after birth) that developed a spontaneous inactivating mutation of a single allele of the VEGFR-3 gene after C3H mice were exposed to a teratogen (ethylnitrosourea). Similar to VEGFR-3 knockout mice, Chy mice with homozygous mutations died in utero. Heterozygous mice usually survived birth and had chylous ascites and chylothorax at birth. In approximately 90% of offspring, this ascites was spontaneously resorbed in the first 3 weeks after birth, and the mice grew normally. However, adult Chy mice had peripheral edema, hypoplastic or absent skin lymphatic vessels (but normal vasculature), adipose deposition, hyperkeratosis, and impaired lymphatic fluid transport capacity. The authors showed that this phenotype could be effectively reversed, and lymphatic vessels could proliferate after adenoviral gene therapy with VEGF-C (resulting in high local concentrations of this growth factor). Rutkowski et al34 used this same model to show that Chy mice have increased dermal collagen and lipid deposition and decreased hydraulic conductivity. Applying a similar although more targeted approach than with the Chy mice, Haiko et al35 produced conditional knockouts of the VEGFR-3 gene. Similar to the Chy mice, heterozygous VEGFR-3 knockout embryos survived and were born with chylous ascites that resolved spontaneously. These mice also had severely atrophic lymphatic vasculature and skin edema. Makinen et al36 used an interesting approach with a soluble form of VEGFR-3 expressed only in the skin. In this model, the VEGF-C and VEGF-D binding domains of VEGFR-3 were coupled with the Fc portion of immunoglobulin G and were expressed constitutively under the keratin promoter. Because keratin was expressed only by keratinocytes, the expression of this soluble receptor (soluble because of the Fc domain)—capable of binding VEGF-C and VEGF-D but not transmitting their signal—was limited to the skin. In these mice, fetal lymphangiogenesis in the skin was severely limited, and the mice developed a primary lymphedema phenotype, with swelling of the feet, adipose deposition, impaired immunity, and dermal fibrosis.

Conclusion The pathogenesis of lymphedema is still unknown. However, the recent development of numerous useful surgical and genetic models of lymphedema has provided important insights into the pathologic mechanisms of this disabling disease. The use of the right model to ask the right questions remains a challenge, and this decision should be based on the availability of reagents, funds, size of the lymphatic channels, and chronicity of lymphedema.

R EFERENCES 1. DiSipio T, Rye S, Newman B, et al. Incidence of unilateral arm lymphoedema after breast cancer: a systematic review and meta-analysis. Lancet Oncol 14:500-515, 2013. 2. Drinker CK, Field ME, Homans J. The experimental production of edema and elephantiasis as a result of lymphatic obstruction. Am J Physiol 108:509-520, 1934. 3. Olszewski W, Machowski Z, Sokolowski J, et al. Experimental lymphedema in dogs. J Cardiovasc Surg (Torino) 9:178-183, 1968. 4. Danese CA, Georgalas-Bertakis M, Morales LE. A model of chronic postsurgical lymphedema in dogs’ limbs. Surgery 64:814-820, 1968.

K23349_Neligan_46_Animal Model_r4_dc_0581-0594.indd 591

5/27/2015 12:58:28 PM

592

Part VI  Research and Future Directions

5. Chen HC, Pribaz JJ, O’Brien BM, et al. Creation of distal canine limb lymphedema. Plast Reconstr Surg 83:1022-1026, 1989. 6. Suami H, Yamashita S, Soto-Miranda MA, et al. Lymphatic territories (lymphosomes) in a canine: an animal model for investigation of postoperative lymphatic alterations. PLoS One 8:e69222, 2013. 7. Tobbia D, Semple J, Baker A, et al. Lymphedema development and lymphatic function following lymph node excision in sheep. J Vasc Res 46:426-434, 2009. 8. Gong-Kang H, Yuan-Pai H. An experimental model for lymphedema in rabbit ear. Microsurgery 4:236242, 1983. 9. Kanter MA, Slavin SA, Kaplan W. An experimental model for chronic lymphedema. Plast Reconstr Surg 85:573-580, 1990. 10. Anthony JP, Foster RD, Price DC, et al. Lymphatic regeneration following microvascular limb replantation: a qualitative and quantitative animal study. J Reconstr Microsurg 13:327-330, 1997. 11. Slavin SA, Van den Abbeele AD, Losken A, et al. Return of lymphatic function after flap transfer for acute lymphedema. Ann Surg 229:421-427, 1999. 12. Rutkowski JM, Moya M, Johannes J, et al. Secondary lymphedema in the mouse tail: lymphatic hyperplasia, VEGF-C upregulation, and the protective role of MMP-9. Microvasc Res 72:161-171, 2006. 13. Clavin NW, Avraham T, Fernandez J, et al. TGF-beta1 is a negative regulator of lymphatic regeneration during wound repair. Am J Physiol Heart Circ Physiol 295:H2113-H2127, 2008. 14. Halsted WS. The swelling of the arm after operation for cancer of the breast—elephantiasis chirugica: its causes and prevention. Bull Johns Hopkins Hosp 32:309-313, 1921. 15. Reichert F. The regeneration of the lymphatics. Arch Surg 13:871, 1926. 16. Tobbia D, Semple J, Baker A, et al. Experimental assessment of autologous lymph node transplantation as treatment of postsurgical lymphedema. Plast Reconstr Surg 124:777-786, 2009. 17. Baker A, Kim H, Semple JL, et al. Experimental assessment of pro-lymphangiogenic growth factors in the treatment of post-surgical lymphedema following lymphadenectomy. Breast Cancer Res 12:R70, 2010. 18. Lahteenvuo M, Honkonen K, Tervala T, et al. Growth factor therapy and autologous lymph node transfer in lymphedema. Circulation 123:613-620, 2011. 19. Honkonen KM, Visuri MT, Tervala TV, et al. Lymph node transfer and perinodal lymphatic growth factor treatment for lymphedema. Ann Surg 257:961-967, 2013. 20. Blum KS, Hadamitzky C, Gratz KF, et al. Effects of autotransplanted lymph node fragments on the lymphatic system in the pig model. Breast Cancer Res Treat 120:59-66, 2010. 21. Fu K, Izquierdo R, Vandevender D, et al. Transplantation of lymph node fragments in a rabbit ear lymphedema model: a new method for restoring the lymphatic pathway. Plast Reconstr Surg 101:134141, 1998. 22. Szuba A, Skobe M, Karkkainen MJ, et al. Therapeutic lymphangiogenesis with human recombinant VEGF-C. FASEB J 16:1985-1987, 2002. 23. Takahashi M, Furukawa T. [Cytological and clinical characteristics of the patients with adult Ph1-positive acute leukemia] Rinsho Ketsueki 32:345-353, 1991. 24. Wang GY, Zhong SZ. A model of experimental lymphedema in rats’ limbs. Microsurgery 6:204-210, 1985. 25. Lee-Donaldson L, Witte MH, Bernas M, et al. Refinement of a rodent model of peripheral lymphedema. Lymphology 32:111-117, 1999. 26. Saito Y, Nakagami H, Morishita R, et al. Transfection of human hepatocyte growth factor gene ameliorates secondary lymphedema via promotion of lymphangiogenesis. Circulation 114:1177-1184, 2006. 27. Mendez U, Stroup EM, Lynch LL, et al. A chronic and latent lymphatic insufficiency follows recovery from acute lymphedema in the rat foreleg. Am J Physiol Heart Circ Physiol 303:H1107-H1113, 2012. 28. Avraham T, Zampell JC, Yan A, et al. Th2 differentiation is necessary for soft tissue fibrosis and lymphatic dysfunction resulting from lymphedema. FASEB J 27:1114-1126, 2013.

K23349_Neligan_46_Animal Model_r4_dc_0581-0594.indd 592

5/27/2015 12:58:28 PM

Chapter 46  Animal Models of Lymphedema

593

29. Zampell JC, Yan A, Elhadad S, et al. CD4(1) cells regulate fibrosis and lymphangiogenesis in response to lymphatic fluid stasis. PLoS One 7:e49940, 2012. 30. Chung J, Kwon Y, Lee T, et al. An experimental study for mouse lymphedema model. Korean J Vasc Endovasc Surg 27:114-119, 2011. 31. Oashi K, Furukawa H, Oyama A, et al. A new model of acquired lymphedema in the mouse hind limb: a preliminary report. Ann Plast Surg 69:565-568, 2012. 32. Zampell JC, Yan A, Avraham T, et al. Temporal and spatial patterns of endogenous danger signal expression after wound healing and in response to lymphedema. Am J Physiol Cell Physiol 300:C1107C1121, 2011. 33. Karkkainen MJ, Saaristo A, Jussila L, et al. A model for gene therapy of human hereditary lymphedema. Proc Natl Acad Sci U S A 98:12677-12682, 2001. 34. Rutkowski JM, Markhus CE, Gyenge CC, et al. Dermal collagen and lipid deposition correlate with tissue swelling and hydraulic conductivity in murine primary lymphedema. Am J Pathol 176:1122-1129, 2010. 35. Haiko P, Makinen T, Keskitalo S, et al. Deletion of vascular endothelial growth factor C (VEGF-C) and VEGF-D is not equivalent to VEGF receptor 3 deletion in mouse embryos. Mol Cell Biol 28:4843-4850, 2008. 36. Makinen T, Jussila L, Veikkola T, et al. Inhibition of lymphangiogenesis with resulting lymphedema in transgenic mice expressing soluble VEGF receptor-3. Nat Med 7:199-205, 2001.

K23349_Neligan_46_Animal Model_r4_dc_0581-0594.indd 593

5/27/2015 12:58:28 PM

K23349_Neligan_46_Animal Model_r4_dc_0581-0594.indd 594

5/27/2015 12:58:28 PM

C hapter 47 Harnessing Stem Cell–Mediated Lymphangiogenesis for the Treatment of Lymphedema

Har the

Ramin Shayan, Geraldine Mitchell, Anthony Penington

K ey P oints • New molecular understanding of the growth and development of the lymphatic system is creating new opportunities for molecular intervention in lymphangiogenesis. • Lymphatic growth in tumors is one area of research that is revealing new insights into lymphatic growth and development, including the fact that tumor-associated macrophages can become lymphatic endothelial cells. • Tissue engineering approaches to the restoration of collecting lymphatics may hold promise for the treatment of lymphedema. • Adipose-derived mesenchymal stem cells appear promising for the treatment of lymphedema. However, their effectiveness, if confirmed, is probably the result of their ability to release growth factors in a sustained manner rather than their ability to differentiate into lymphatic tissue components.

Recent advances in the molecular biology, visualization techniques, and genetic modeling of the lymphatics have shed light on the developmental origins of this intricate vascular system.1,2 This in turn has created new opportunities for the manipulation of lymphatic biology for clinical benefit, including the exploitation of protein growth factors, small molecules, and neutralizing antibodies to target the lymphatic vasculature.3 In 2002 Niklason et al4 stated that tissue engineering approaches for lymphatic engineering were “currently nonexistent.” Since then, several approaches, including the use of lymphangiogenic growth factors, lymphatic grafts, tissue transfers, and mesenchymal stem cells (MSCs), have largely been used experimentally and occasionally clinically to regenerate lymphatic vessels and their networks for the treatment of secondary lymphedema. The engineering of new tissues, including new lymphatics, requires a combination of cells, matrix, and growth factors. MSCs and in particular adipose-derived stem cells (ADSCs) are a promising source of cells for lymphatic regeneration, although more recent evidence indicates that their real value may be in their ability to sustainably release a range of growth factors.5 595

K23349_Neligan_47_Harnessing Stem_r4_595-605.indd 595

5/27/2015 1:01:49 PM

596

Part VI  Research and Future Directions

Cellular Origin of Lymphatic Endothelial Cells Lymphatic endothelial cells (LECs) are the most important cell type in lymphatic regeneration and therefore the key to any restoration of lymphatic function in lymphedema. The earliest LECs in embryonic development arise from primitive venous endothelial cell precursors2,6 in the anterior cardinal veins and a separate superficial venous plexus.7-9 The initial sprouting of the primitive lymphatic vessels is preempted by a transition from venous to lymphatic endothelial expression profiles of distinct LEC markers.9 In murine models, Sry-related HMG box containing-18 (SOX18) and chicken ovalbumin upstream promoter or NR2F2 (COUP-TFII) expressing cells cluster into a prelymphatic subsection of the veins from 14.5 days to postcoitus.7 The resulting jugulodigastric lymphatic sacs then give rise to collecting lymphatics, whereas the more peripherally derived LECs, which form the more superficial subtypes of lymphatics, continue to expand and only mature during later developmental stages in some areas postpartum.3,8 Therefore endothelial cells from the initial and collecting lymphatics have a distinct origin and biology, and both are required for a functioning lymphatic system.8 Distal wing bud grafting experiments of chick into quail embryos have been shown to generate lymphatics comprised of both chick and quail LECs. This finding suggests that wing bud lymphatics do not exclusively develop from sprouts from nearby lymph sacs but also develop by recruiting circulating “lymphangioblasts.”7 Macrophages or circulating endothelial progenitor cells have also been proposed as a potential nonendothelial LEC progenitor.8,10 Macrophages have been observed to promote angiogenesis and act as potential sources of LECs in tumor models.10 These tumor-associated macrophages have been shown to acquire LEC-specific markers, such as prospero homeobox 1 (PROX1), vascular endothelial growth factor receptor 3 (VEGFR-3), lymphatic vessel endothelial hyaluronan receptor 1 (LYVE1), and podoplanin while simultaneously undergoing observable downregulation of myeloid markers.7 Such “reprogramming” of myeloid into LEC identity suggests that tumor-associated macrophages and possibly other macrophages may contribute to the lymphatic vasculature,8 although studies in developing mouse embryos have only shown that macrophages contribute to the modulation of lymphatic vessel caliber by regulating LEC proliferation without transdifferentiation or incorporation into vessels.8 A key feature of embryonic lymphangiogenesis is venous transdifferentiation into LECs.11 The maintenance of LEC identity requires ongoing PROX1 expression, without which reversal to a blood endothelial phenotype occurs.12,13 In contrast, negative regulatory control of lymphangiogenesis is mediated by highly preserved mechanisms involving Notch, CYP26B1 (a retinoic acid degradation enzyme), and transforming growth factor-beta (TGF-beta)/bone morphogenic protein (BMP) signaling pathways.14,15 During development, Notch expression (particularly NOTCH1) is integral to restricting the transition to LEC fate to certain areas of precursor veins.16,17 During adulthood, another protein growth factor commonly implicated in fibrosis, TGF-beta inhibits LEC proliferation,10,18 as have BMP2, BMP4, and BMP.10 TGF-beta is highly expressed in scar tissue and therefore may have a role in the prevention of lymphatic regeneration after surgery.5,18 Once established, lymphatic vessels may undergo significant remodeling controlled by expression of key molecular markers with specificity for LECs, such as VEGFR-3, LYVE1, podoplanin, and NRP2, which allow LECs to acquire responsiveness to protein growth factors that are vital to forming and maturing a vascular plexus.3,7,10 The critical signaling pathways determining LEC

K23349_Neligan_47_Harnessing Stem_r4_595-605.indd 596

5/27/2015 1:01:49 PM

Chapter 47  Harnessing Stem Cell–Mediated Lymphangiogenesis for the Treatment of Lymphedema

597

remodeling revolve around the vascular endothelial growth factor C (VEGF-C)/collagen and calcium-binding EGF domain-containing protein1 (CCBE1)/VEGFR-3 pathway, a signaling axis that promotes LEC migration, proliferation, and survival and thus is indispensable for both embryonic and adult lymphangiogenesis.7 VEGF-C loss of function experiments reveal that an absence of this key growth factor results in unaltered LEC specification, because the embryos fail to form a complete lymphatic vasculature as a result of defective lymphatic sprouting. Furthermore, VEGF-C was not active in all the areas of the embryos in which it was expressed, because its ability to activate VEGFR-3 relies on VEGF-C modulation by CCBE1, which processes VEGF-C by means of a disintegrin and metalloproteinase with a thrombospondin motif (ADAMTS3)– dependent mechanism.7,10

Tumor-Induced Lymphangiogenesis: Lessons for Lymphatic Remodeling Tumors spread either by hematogenous or lymphatic spread.3,7 The formation of lymphatics in both human tumors and animal models has been linked to enhanced lymphatic metastasis. Therefore the process of lymphangiogenesis has also become a target for therapeutic restriction of tumor metastasis.3,7,19 The intense research in this area has created new knowledge regarding lymphatic growth and remodeling. Although primarily directed at inhibiting lymphatic growth, it may also generate new insights into how lymphatic regeneration may be achieved. An important mechanism contributing to tumor-induced lymphangiogenesis is outsprouting from vessels in and around a primary tumor mass from preexisting initial lymphatics.19 As this occurs in response to stimuli that may include VEGF-C, vascular endothelial growth factor D (VEGF-D), plateletderived growth factor subunit beta (PDGFB), fibroblast growth factor 2 (FGF2), and angiopoietin,7 each of these signaling molecules has consequently become considered potential targets for molecular therapeutics to restrict tumor-induced lymphangiogenesis.3 The same molecules may also be targets for lymphatic regeneration.20 Small-caliber initial lymphatic vessels and larger collecting vessels that make up the lymphatic system differ in their morphology, structure, and anatomic locations and perform distinct specialized functions that contribute to their respective normal physiologic roles.3,7,19 Most research investigating mechanisms behind lymphogenous metastasis of cancer cells has focused on elucidating the influences of lymphangiogenic growth factors on the initial lymphatic vessel subtypes within or around a primary tumor,19,21 whereas the collecting lymphatic vessels that drain the tissues of the tumor toward the regional lymph nodes have remained largely ignored.22 Whereas ambient smaller lymphatics largely respond to lymphangiogenic factors by proliferation and/or sprouting to generate tumor-associated lymphangiogenesis, the larger collecting vessels respond to the same lymphangiogenic stimuli in a unique and quite distinctive manner.22-25 Far from passive conduits of metastatic cells, more recent findings suggest that collecting lymphatic vessels undergo a significant remodeling of their own, which contributes critically to the process of tumor spread22,23 (Fig. 47-1). Observations of murine models of VEGF-C-overexpressing metastatic tumors demonstrated that drainage from the primary tumor by the collecting lymphatic performs an active role in enhancing tumor dissemination through increasing fluid flow by dilating, an increase in collecting lymphatic diameter attributed to LEC proliferation.22-26 Similarly, it was shown that VEGF-D secreted by a flank xenograft tumor model also induced dilation of the collecting

K23349_Neligan_47_Harnessing Stem_r4_595-605.indd 597

5/27/2015 1:01:49 PM

598

Part VI  Research and Future Directions

Adipose-derived stem cell (ADSC)

Angiogenesis VEGF family of growth factors (e.g., VEGF-D)

VEGF-independent growth factors (e.g., IL-8, IGF-1)

ADSC secretion profile

Suppression of immune and inflammatory responses Reduction of fibrogenesis

VEGFR-3

Non-VEGF receptor Lymphatic endothelial cell

Fibrosis Increased proliferation Increased migration

Increased lymphangiogenesis

Increased tube formation

Lymphedema

Infection

Enhanced lymphatic function

FIG. 47-1  Suggested mechanism of ADSC-mediated cellular repair of lymphedema and prevention of the resulting fibrotic cascade.

lymphatics draining from the primary tumor to the axilla, which was critical for cancer spread to the sentinel lymph node.22 However, in contrast to the VEGF-C model and the effect of VEGF-D on the initial lymphatics,6 the mechanism by which circumferential dilation occurs is not by endothelial proliferation but through specific prostaglandin-mediated responses to VEGF-D.22 Treatment with nonsteroidal antiinflammatory drugs, including aspirin, not only reverses the VEGF-D-driven morphologic remodeling of collecting lymphatics but also reduces the rate of tumor metastasis to draining lymph nodes and distant organs.22 These studies illustrate the complexity of lymphangiogenic signaling and that molecular stimulation of lymphangiogenesis is context specific and modulated by other growth factors and drug treatment. They also highlight the need to consider the growth and maintenance of collecting lymphatics and not just the initial lymphatics in lymphedema treatment.7

Use of Growth Factors to Stimulate Lymphangiogenesis The best-studied lymphangiogenic growth factors are VEGF-C and VEGF-D and their receptor, VEGFR-3.7,27 VEGF-C delivery in several forms, including recombinant protein, viral vector, or naked plasmid, can reverse surgically induced lymphedema in numerous animal species with dif-

K23349_Neligan_47_Harnessing Stem_r5_595-605.indd 598

5/29/2015 12:24:33 PM

Chapter 47  Harnessing Stem Cell–Mediated Lymphangiogenesis for the Treatment of Lymphedema

599

ferent lymphedema models.7 However, problems are associated with VEGF-C therapy, because it may initially increase LEC proliferation and lymphatic vessel growth. VEGF-C does not improve LEC migration or functionality, and VEGF-C alone cannot permanently improve lymphatic size, density, or organization in regenerating adult skin.28 More recent studies have targeted growth factors outside the VEGF family. In one important study, interleukin 8 (IL-8) was used in the treatment of a surgical lymphedema model with a murine tail.29 The authors showed that IL-8 promoted LEC proliferation, tube formation, and migration without activating either VEGFR-3 signaling or the inflammatory pathways normally associated with IL-8 activity. In addition to different growth factors, numerous other factors probably influence lymphangiogenesis, in particular fluid flow. It is known that lymphatic growth occurs in the direction of lymphatic fluid flow30 and that interstitial fluid flow is essential for lymphatic capillary regeneration in skin.28 This suggests that the combination of growth factor treatment with physical therapies may be more effective than growth factors alone in the treatment of lymphedema.

Tissue Engineering Several studies have attempted tissue engineering of lymphatic vessel networks in vitro, but few of these techniques have been used in lymphedema models. Lymphatic vessel networks are known to grow in human extracellular dermal matrix scaffolds in vivo.31 Tissue engineering of lymphatic networks has been achieved with human LEC multicellular spheroid implantation into a Matrigel/ fibrin matrix in mice,32 growing human lymphatic capillary networks in three-dimensional hydrogels and transplanting these scaffolds into nude rats. These studies showed inosculation (joining) of the engineered networks to the lymphatic vessels of the recipient skin sites and draining of tissue fluids through the implanted lymphatic vessels. The generation of a lymphatic endothelialized–tissue engineered macrograft based on a synthetic scaffold33 has been attempted; however, the lymphatic grafts were implanted subdermally but not tested functionally within the lymphatic vascular system.

Mesenchymal Stem Cells Despite the explosion of interest in MSC therapies in the past 10 years,5 stem cell treatment for lymphedema has only been seriously examined recently.34 MSCs can be isolated from various tissues in adults and have a pluripotent potential, which means they can be induced to differentiate into cell types of bone, cartilage, and fat and more recently those of other tissues, such as liver, skeletal muscle, and cardiac muscle.5 Adipose tissue is known as one of the richest sources of stem cells.35,36 This, along with the relative ease of accessing adipose tissue by liposuction in a single-stage procedure5 and their isolation with commercially available automated isolation equipment36 that is both rapid and safe to use, has made adipose-derived MSCs (ADSCs) an increasingly popular stem cell option.5 Fat grafting to treat radiation-induced soft tissue injury was initially described5 as an observation37 and then replicated in animal models,38 and lymphedema is a significant factor in this scenario.4 The presence of ADSCs in fat grafts is a key component of this treatment.5,34,38 The clinical benefits of fat grafting have been attributed to the multipotent differentiation potential of ADSCs,

K23349_Neligan_47_Harnessing Stem_r4_595-605.indd 599

5/27/2015 1:01:49 PM

600

Part VI  Research and Future Directions

but it is increasingly recognized that the more important consideration is the role that ADSCs play in adipogenesis and angiogenesis while modulating inflammation and immunity.5,34-36 Recent studies have found that ADSCs have an extensive secretory profile consisting of cytokines, chemokines, and growth factors.5,39,40 Although it was previously thought that ADSCs differentiate to replace injured cells (“host replacement” or “building block” repair theories), it is now understood that secreted paracrine mediators elicit effects on surrounding tissues, which may be much more important (see Chapter 4). The particular secretory profile of ADSCs suggests that they specifically influence molecular and biologic pathways of tissue regeneration, angiogenesis, and lymphangiogenesis while suppressing local immune and inflammatory responses and reducing fibrogenesis.5,38,41,42

Evidence of Lymphangiogenic Properties of Adipose-Derived Stem Cells In vitro studies have modeled scratch wound closure by cultured murine fibroblasts and immortalized keratinocytes separately and in co-culture with stem cell condition media.43 Detection of levels of interleukin 6 (IL-6), IL-8, monocyte chemotactic protein 1 (MCP-1), regulated on activation normal T cell expressed and secreted (RANTES), and TGF-beta 1 expression suggested that paracrine signaling mediated promigratory effects of stem cell condition media.44 In particular, the profile of ADSC paracrine secretion as detected on mRNA mini-arrays indicates that ADSCs produce a growth factor secretion profile that is distinct from other varieties of stem cells, with particularly high expression levels of IL-8, insulin-like growth factor 1, and VEGF-D proteins.39 These three growth factors have demonstrated lymphangiogenic activity and suggest a privileged role for ADSCs in the process of lymphangiogenesis.5 Wounded and irradiated tissues can often demonstrate significant lymphatic fluid stasis, clinically similar to lymphedema.45 This lymphatic fluid stasis induces the expression of TGF-beta 1 in irradiated tissues. The studies of Rigotti et al37 and Avraham et al41 suggest that TGF-beta probably exerts an antilymphangiogenic effect. Further studies have shown that blocking TGF-beta 1 with simultaneous in vitro stimulation with VEGF-C leads to enhanced survival rates of implanted ADSCs.5,41 This approach also boosted ADSC expression levels of potentially lymphangiogenic proteins VEGF-A, VEGF-C, and the LEC markers podoplanin and Prox1.5,40-42 Collectively, these data indicate that both the baseline expression of lymphangiogenic growth factors (IL-8, insulin-like growth factor 1, and VEGF-D) produced by ADSCs, in addition to the range of lymphangiogenic factors produced by ADSCs when stimulated by particular influences, may be able to facilitate lymphangiogenesis to potentially aid in the reduction or reversal of lymphedema.5,39-42 Overall, further detailed systematic analysis of the secretory expression profile is required to identify which specific growth factors are released in vivo and how they may modulate the processes involved in wound healing, angiogenesis, and lymphangiogenesis.5,39,44

K23349_Neligan_47_Harnessing Stem_r4_595-605.indd 600

5/27/2015 1:01:49 PM

Chapter 47  Harnessing Stem Cell–Mediated Lymphangiogenesis for the Treatment of Lymphedema

601

Treatment of Lymphedema With Adipose-Derived Stem Cells in Animals and Humans Some benefits of ADSC therapy have been found in animal models of lymphedema. Yan et al42 found that transient short-term stimulation of ADSCs with VEGF-C resulted in enhanced VEGF-A, VEGF-C, and Prox1 expression in vitro and was associated with a marked increase in prolymphangiogenic response after in vivo implantation in an animal model.45 The stimu­ lated cells also showed increased cellular proliferation and survival capacity, and this lymphangiogenic response was potentiated by TGF-beta 1 blockade.42 Shimizu et al46 injected 2 million mouse ADSCs into a mouse tail lymphedema model at day 1 and assessed tail circumference until day 29. Lymphedema improved significantly by local injection of ADSCs, and histologic analysis indicated that lymphatic capillary density was greater in the ADSC group than in controls. Hwang et al47 used a hindlimb lymphedema model in mice and implanted human ADSCs or a VEGF-C-containing hydrogel or a combination of both and followed the animals for 28 days. The combined treatment showed a significant decrease in dermal edema depth compared with the groups of mice that received human adipose-derived stem cells (hADSCs) only or VEGF-C hydrogel only. Immunohistochemical analysis showed that the combined treatment group had significantly greater lymphatic vessel regeneration than all the other groups.47 The hADSCs were detected in the implantation sites of all mice in the hADSC/VEGF-C group and showed a lymphatic endothelial differentiation in prelabeled implanted hADSCs, which were LYVE1 positive.47 The long-term efficacy of these studies has been questioned.48 However, Zhou et al49 used a surgical and radiotherapy model in rabbit hindlimbs and followed the injured limbs for 6 months. Three months after the development of lymphedema, rabbit bone marrow MSCs (10 million) and/ or VEGF-C protein (150 ng/kg) were injected into the lymphedematous limbs at the site of skin removal and irradiation. An increase in lymphatic vessel numbers was evident 28 days after treatment in all treatment groups compared with untreated controls, and 6 months after surgery and radiotherapy, there were significant decreases in limb volume.49 The combination treatment was superior to experimental groups that used the individual treatments alone. Further animal studies are clearly necessary to assess the efficacy of stem cell therapy, but early results are promising.49 Two small human studies of ADSC treatment for lymphedema after axillary dissection for breast cancer have been published.34,50 Both studies reported promising results, with improvement in symptoms and reduction in swelling, but the numbers were small and both studies have been criticized on methodologic grounds.48 Larger clinical studies will be needed to assess the effectiveness of ADSC therapy for lymphedema.

Future Approaches to the Treatment of Lymphedema The molecular processes surrounding lymphangiogenesis (the same processes involved in developmental lymphedema) are becoming better understood, in particular because of interest in the role of the lymphatic system in the spread of cancer. Overall lymphedema resulting from

K23349_Neligan_47_Harnessing Stem_r4_595-605.indd 601

5/27/2015 1:01:49 PM

602

Part VI  Research and Future Directions

oncologic surgery is far more common. However, similar genetic derangements may also have an influence that make some individuals more susceptible to secondary lymphedema. Regardless, more detailed knowledge of the mechanisms governing lymphatic biology is required to safely design treatments that stimulate lymphatic growth, particularly in patients who have developed lymphedema after previous cancer treatment. To date, most novel strategies to manipulate the lymphatic system (either to restrict tumor lymphatics and therefore metastasis or to promote lymphangiogenesis in lymphedema or wound healing) have focused on the VEGF-C/VEGFR-3 signaling axis. Preclinical data in cancer treatment suggest a benefit in targeting tumor-induced lymphangiogenesis to reduce solid tumor metastases.3,7 Although clinical studies of tumor sections correlate enhanced lymphatic vessel density with higher rates of metastasis, actual clinical trials restricting the growth of lymphatic vessels in human cancer have yet to be fully evaluated.3,7 The two clinical trials performed to target the VEGF-C/VEGFR-3 pathway have been combined with antiangiogenic molecules. The first combined the prototype anti-VEGF-A monoclonal antibody bevacizumab (Avastin) with VGX-100 (anti-VEGF-C neutralizing antibody, NCT01514123). Meanwhile the other trial, which targeted VEGFR-3 with anti-VEGFR-3 monoclonal antibody (NCT01288989) in subjects with advanced solid tumors refractory to standard therapy or for whom no standard therapy was available has completed the phase 1 clinical trial at the time of this writing. Tissue engineering therapies for the treatment of lymphedema are promising, but the ability of engineered constructs to become incorporated into existing lymphatic networks requires more study. In addition, understanding that collecting lymphatics are actively remodeled provides both a challenge to tissue engineering and an opportunity for lymphatic regeneration. The identification of bone marrow–derived LEC progenitor cells has suggested that the capacity to regenerate lymphatic function may involve complex interactions between prochemotactic signaling cascades and those signals that preserve stem cell populations in their respective sites of origin. Overall, further exploration of the developmental pathways that govern lymphangiogenesis is essential to expand the identification, characterization, and optimization of molecular targets so that new directions in treatments for lymphedema can be devised. Although promising work involving the VEGF-C/VEGF-D/VEGFR-3 axis has suggested some potentially mechanistic­ ally sound approaches, the introduction of systemic treatment with purified protein or adenoviral vector is some considerable time away. In the meantime, ADSCs show a particular predi­ lection for the secretion of lymphangiogenic growth factors (both VEGFR-3-dependent and VEGFR-3-independent) and may therefore provide the perfect vehicle by which to deliver these chemokine mediators, which are generated from an autologous source and at minimal expense for the treatment of primary and/or secondary human lymphedema.

K23349_Neligan_47_Harnessing Stem_r4_595-605.indd 602

5/27/2015 1:01:49 PM

Chapter 47  Harnessing Stem Cell–Mediated Lymphangiogenesis for the Treatment of Lymphedema

603

C linical P earls • ADSCs are relatively abundant and are easy to obtain and isolate from patient lipoaspirate. • The secretory profile of ADSCs may particularly favor lymphangiogenesis compared with other MSC populations. • Surviving ADSC populations in injected fat grafts may function as persistent sources of protein growth factors that improve healing and localized lymphedema after cancer therapy. • Larger, randomized clinical trials evaluating ADSC therapy in lymphedema are needed.

R EFERENCES 1. Huntington GS, McClure CFW. The anatomy and development of the jugular lymph sac in the domestic cat (Felis domestica). Am J Anat 10:177-311, 1910. 2. Sabin FR. On the origin of the lymphatic system from the veins, and the development of the lymph hearts and thoracic duct in the pig. Am J Anat 1:367-389, 1902. 3. Shayan R, Achen MG, Stacker SA. Lymphatic vessels in cancer metastasis: bridging the gaps. Carcinogenesis 27:1729-1738, 2006. 4. Niklason LE, Koh J, Solan A. Tissue engineering of the lymphatic system. Ann N Y Acad Sci 979:2734; discussion 35-38, 2002. 5. Shukla L, Morrison WA, Shayan R. Adipose-derived stem cells in radiotherapy injury: a new frontier. Front Plast Surg 2:1, 2015. 6. Wigle JT, Oliver G. Prox1 function is required for the development of the murine lymphatic system. Cell 98:769-778, 1999. 7. Stacker SA, Williams SP, Karnezis T, et al. Lymphangiogenesis and lymphatic vessel remodelling in cancer. Nat Rev Cancer 14:159-172, 2014. 8. Francois M, Short K, Secker GA, et al. Segmental territories along the cardinal veins generate lymph sacs via a ballooning mechanism during embryonic lymphangiogenesis in mice. Dev Biol 364:89-98, 2012. 9. Srinivasan RS, Dillard ME, Lagutin OV, et al. Lineage tracing demonstrates the venous origin of the mammalian lymphatic vasculature. Genes Dev 21:2422-2432, 2007. 10. Francois M, Karnezis T, Shayan R. Ordered chaos: harnessing developmental pathways in tumorinduced lymphangiogenesis. J Clin Cell Immunol 5:270, 2014. 11. Francois M, Caprini A, Hosking B, et al. Sox18 induces development of the lymphatic vasculature in mice. Nature 456:643-647, 2008. 12. Srinivasan RS, Oliver G. Prox1 dosage controls the number of lymphatic endothelial cell progenitors and the formation of the lymphovenous valves. Genes Dev 25:2187-2197, 2011. 13. Johnson NC, Dillard ME, Baluk P, et al. Lymphatic endothelial cell identity is reversible and its maintenance requires Prox1 activity. Genes Dev 22:3282-3291, 2008. 14. Bowles J, Secker G, Nguyen C, et al. Control of retinoid levels by CYP26B1 is important for lymphatic vascular development in the mouse embryo. Dev Biol 386:25-33, 2014.

K23349_Neligan_47_Harnessing Stem_r4_595-605.indd 603

5/27/2015 1:01:50 PM

604

Part VI  Research and Future Directions

15. Marino D, Dabouras V, Brandli AW, et al. A role for all-trans-retinoic acid in the early steps of lymphatic vasculature development. J Vasc Res 48:236-251, 2011. 16. Fatima A, Culver A, Culver F, et al. Murine Notch1 is required for lymphatic vascular morphogenesis during development. Dev Dyn 243:957-964, 2014. 17. Geudens I, Herpers R, Hermans K, et al. Role of delta-like-4/Notch in the formation and wiring of the lymphatic network in zebrafish. Arterioscler Thromb Vasc Biol 30:1695-1702, 2010. 18. James JM, Nalbandian A, Mukouyama YS. TGFb signaling is required for sprouting lymphangiogenesis during lymphatic network development in the skin. Development 140:3903-3914, 2013. 19. Shayan R, Inder R, Karnezis T, et al. Tumor location and nature of lymphatic vessels are key determinants of cancer metastasis. Clin Exp Metastasis 30:345-356, 2013. 20. Shayan R, Karnezis T, Tsantikos E, et al. A system for quantifying the patterning of the lymphatic vasculature. Growth Factors 25:417-425, 2007. 21. Wick N, Haluza D, Gurnhofer E, et al. Lymphatic precollectors contain a novel, specialized subpopulation of podoplanin low, CCL27-expressing lymphatic endothelial cells. Am J Pathol 173:1202-1209, 2008. 22. Karnezis T, Shayan R, Caesar C, et al. VEGF-D promotes tumor metastasis by regulating prostaglandins produced by the collecting lymphatic endothelium. Cancer Cell 21:181-195, 2012. 23. He Y, Rajantie I, Pajusola K, et al. Vascular endothelial cell growth factor receptor 3-mediated activation of lymphatic endothelium is crucial for tumor cell entry and spread via lymphatic vessels. Cancer Res 65:4739-4746, 2005. 24. Hoshida T, Isaka N, Hagendoorn J, et al. Imaging steps of lymphatic metastasis reveals that vascular endothelial growth factor-C increases metastasis by increasing delivery of cancer cells to lymph nodes: therapeutic implications. Cancer Res 66:8065-8075, 2006. 25. Wirzenius M, Tammela T, Uutela M, et al. Distinct vascular endothelial growth factor signals for lymphatic vessel enlargement and sprouting. J Exp Med 204:1431-1440, 2007. 26. Paquet-Fifield S, Levy SM, Sato T, et al. Vascular endothelial growth factor-d modulates caliber and function of initial lymphatics in the dermis. J Invest Dermatol 133:2074-2084, 2013. 27. Partanen TA, Alitalo K, Miettinen M. Lack of lymphatic vascular specificity of vascular endothelial growth factor receptor 3 in 185 vascular tumors. Cancer 86:2406-2412, 1999. 28. Goldman J, Le TX, Skobe M, et al. Overexpression of VEGF-C causes transient lymphatic hyperplasia but not increased lymphangiogenesis in regenerating skin. Circ Res 96:1193-1199, 2005. 29. Choi I, Lee YS, Chung HK, et al. IL-8 reduces post-surgical lymphoedema formation by promoting lymphatic vessel regeneration. Angiogenesis 16:29-44, 2013. 30. Planas-Paz L, Lammert E. Mechanical forces in lymphatic vascular development and disease. Cell Mol Life Sci 70:4341-4354, 2013. 31. Swartz MA, Lund AW. Lymphatic and interstitial flow in the tumour microenvironment: linking mechanobiology with immunity. Nat Rev Cancer 12:210-219, 2012. 32. Alajati A, Laib AM, Weber H, et al. Spheroid-based engineering of a human vasculature in mice. Nat Methods 5:439-445, 2008. 33. Marino D, Luginbühl J, Scola S, et al. Bioengineering dermo-epidermal skin grafts with blood and lymphatic capillaries. Sci Transl Med 6:221, 2014. 34. Maldonado GE, Pérez CA, Covarrubias EE, et al. Autologous stem cells for the treatment of postmastectomy lymphoedema: a pilot study. Cytotherapy 13:1249-1255, 2011. 35. Zuk PA, Zhu M, Ashjian P, et al. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell 13:4279-4295, 2002. 36. Ross R, Shayan R, Mutimer KL, et al. Autologous fat graft survival: current state of the art. Ann Plast Surg 73:352-357, 2014. 37. Rigotti G, Marchi A, Galiè M, et al. Clinical treatment of radiotherapy tissue damage by lipoaspirate transplant: a healing process mediated by adipose-derived adult stem cells. Plast Reconstr Surg 119:14091422, 2007.

K23349_Neligan_47_Harnessing Stem_r4_595-605.indd 604

5/27/2015 1:01:50 PM

Chapter 47  Harnessing Stem Cell–Mediated Lymphangiogenesis for the Treatment of Lymphedema

605

38. Coleman CN. Effects of radiation on normal tissue: consequences and mechanisms. Science 304:693694, 2004. 39. Hsiao ST, Asgari A, Lokmic Z, et al. Comparative analysis of paracrine factor expression in human adult MSCs derived from bone marrow, adipose, and dermal tissue. Stem Cells Dev 21:2189-2203, 2012. 40. Takeda K, Sowa Y, Nishino K, et al. ADSCs promote proliferation, migration, and tube formation of LECs in vitro by secreting lymphangiogenic factors. Ann Plast Surg. 2014 Jan 7. [Epub ahead of print] 41. Avraham T, Yan A, Zampell JC, et al. Radiation therapy causes loss of dermal lymphatics and interferes with function by TGF-beta1-mediated tissue fibrosis. Am J Physiol Cell Physiol 299:C589-C605, 2010. 42. Yan A, Avraham T, Zampell JC, et al. Adipose-derived stem cells promote lymphangiogenesis in response to VEGF-C stimulation or TGF-b1 inhibition. Future Oncol 7:1457-1473, 2011. 43. Collawn SS, Banerjee NS, de la Torre J, et al. Adipose-derived stromal cells accelerate wound healing in an organtotypic raft culture model. Ann Plast Surg 68:501-514, 2012. 44. Caplan AI, Dennis JE. Mesenchymal stem cells as trophic mediators. J Cell Biochem 98:1076-1084, 2006. 45. DiSipio T, Rye S, Newman B, et al. Incidence of unilateral arm lymphoedema after breast cancer: a systematic review and meta-analysis. Lancet Oncol 14:500-515, 2013. 46. Shimizu Y, Shibata R, Shintani S, et al. Therapeutic lymphangiogenesis with implantation of adiposederived regenerative cells. J Am Heart Assoc 1:e000877, 2012. 47. Hwang JH, Kim IG, Lee JY, et al. Therapeutic lymphangiogenesis using stem cell and VEGF-C hydrogel. Biomaterials 32:4415-4423, 2011. 48. Toyserkani NM, Christensen ML, Sheikh SP, et al. Stem cells show promising results for lymphoedema treatment—a literature review. J Plast Surg Hand Surg 49:65-71, 2015. 49. Zhou H, Wang M, Hou C, et al. Exogenous VEGF-C augments the efficacy of therapeutic lymphangiogenesis induced by allogenic bone marrow stromal cells in a rabbit model of limb secondary lymphoedema. Jpn J Clin Oncol 41:841-846, 2011. 50. Hou C, Wu X, Jin X. Autologous bone marrow stromal cells transplantation for the treatment of secondary arm lymphoedema: a prospective controlled study in patients with breast cancer related lymphoedema. Jpn J Clin Oncol 38:670-674, 2008.

K23349_Neligan_47_Harnessing Stem_r4_595-605.indd 605

5/27/2015 1:01:50 PM

K23349_Neligan_47_Harnessing Stem_r4_595-605.indd 606

5/27/2015 1:01:50 PM

C hapter 48 Establishment of a Lymphedema Framework Christine Moffatt, Susie Murray

K ey P oints The objectives of the International Lymphoedema Framework charity are as follows: • To raise the national and international profile of lymphedema • To make lymphedema and its management priorities on the national health care agenda

Est

• To enable clinicians to lobby for the appropriate financing and reimbursement of lymphedema care • To address issues of inequity of provision of care • To implement and evaluate lymphedema services based on best practice • To create an international lymphedema community that collectively strives to improve the evidence base for treatment and professional practice • To improve the lives of patients with lymphedema and their families worldwide

The International Lymphoedema Framework (ILF) was established in 2009 as a United Kingdom charity. Its goal is to improve the management of lymphedema and related disorders worldwide by sharing expertise and resources and supporting the efforts in individual countries to develop a long-term strategy for lymphedema. The ILF began in the United Kingdom when it was recognized that a new approach was needed to collectively engage stakeholders to improve access to care and the standard of care for patients who had lymphedema or were at risk of developing the disease. Despite recent research indicating that the condition is common, affecting 4 per 1000 of the general population and nearly 30 per 1000 in the very elderly,1 there is little guidance or dedicated care provision except in sporadic services. This situation is mirrored in many other parts of the world. One of the difficulties is in the description of lymphedema, which has historically referred 607

K23346_Neligan_48_How to Estab_r2_dc_0607-0616.indd 607

5/26/2015 10:14:53 AM

608

Part VI  Research and Future Directions

only to those patients with edema caused by lymphatic failure. There is a growing appreciation that the term chronic edema is more inclusive, because it encompasses those patients with conditions such as venous disease, long-term disability, and systemic causes of edema.1 In all these situations, edema may become an overwhelming sign, and consequently the lymphatic circulation will be overwhelmed and ultimately fail. The aging population, reduced mobility of many older persons, and obesity epidemic are linked to a projected increase in the number of people with the condition. In many parts of the world, such as India2 and Africa,3 lymphedema is predominantly caused by lymphatic filariasis in mosquito-endemic areas.4 Improvements in drugs to stop the transmission of the disease have led to the expectation that the condition will eventually be eradicated.5 However, many patients have significant morbidity, and many have other forms of lymphedema. Despite all the available information, there are still doubts about the true incidence and prevalence of lymphedema, specifically and generally. An international epidemiology study based on an electronic platform is currently running through the ILF to address this lack of robust epidemiology. It uses a common methodology to assess the size and impact of the condition in both developed and developing countries. In 2014 the methodology was developed and validated through three projects running concurrently in the United Kingdom, Denmark, and Japan. Other countries will be invited in the near future to participate in the study. The outcomes of this important project are an international dataset that will indicate the level of neglect regarding lymphedemas and the social, financial, and health importance of governments addressing lymphedema as a public health issue.

What Is the International Lymphoedema Framework? The philosophy of the ILF is based on the belief that the partnership of all stakeholders is vital to changing the landscape of lymphedema management. This partnership includes patients (who play a role at all levels of the organization, including representation at the board level), clinicians, researchers, other national and international lymphedema organizations, and industry. The latter is considered a core member within the partnership and brings a unique expertise and ability to develop innovative responses to clinical challenges. For many professionals, an approach that involves the inclusion of industry is perceived as challenging, and thus there are strict codes of conduct to avoid bias. The international network of expertise that forms the ILF is an invaluable resource that allows projects to have both a national and international dimension and ensures recommendations that address the myriad contextual issues that have an impact on lymphedema treatment.

Structure of the International Lymphoedema Framework The ILF is supported by an elected board of directors that includes the heads of some of the national lymphedema frameworks. These directors have monthly teleconferences (or more often

K23346_Neligan_48_How to Estab_r2_dc_0607-0616.indd 608

5/26/2015 10:14:53 AM

Chapter 48  Establishment of a Lymphedema Framework

609

if other important issues must be addressed). They meet face to face at the ILF conferences; the most recent conferences have been in Glasgow (2014), Montpellier (2012), and Toronto (2011). The 2016 meeting to be held in Darwin, Australia will focus on the Australasia and Pacific regions. An international advisory board is available for advice and expertise, and the members of this board meet during the conference. The members of the board are drawn from developed and developing countries. The ILF welcomes involvement with key agencies and national and international organizations. Current partnerships include the following: • World Health Organization (WHO) • World Alliance for Wound and Lymphedema Care (WAWLC) • Global Alliance for the Elimination of Lymphatic Filariasis (GAELF) • Handicap International (an international charity) • International and national lymphology and wound groups • Patient support groups

What Is a National Lymphedema Framework? A national lymphedema framework is a partnership of stakeholders within a given country who are dedicated to improving lymphedema care in their country. The first national lymphedema framework was established in the United Kingdom in 2002. Through its partnership with those within the field of lymphedema, it developed a national research project, which involved the implementation of an internationally agreed standard of best practice within different service models.6 This led to the adoption of a new classification of lymphedema through the United Kingdom drug tariff, which allowed access to prescriptions for all products required for treatment. As of 2014, the ILF recognized national frameworks in Australia, Belgium, the Netherlands, Denmark, Canada, France, Japan, and the United States. Many leaders of lymphedema groups in other countries (for example, the People’s Republic of China, Taiwan and Singapore) have expressed an interest in becoming members.

Objective of a National Lymphedema Framework The objective of a national framework is to improve the management of lymphedema within that country by developing a strategic plan to enhance care. This plan should include the following: • Assessment of the current care provision, examining both the positives and deficits • Identification of problems of access and payment of care • Involvement of stakeholders in the strategic planning • Adaptation of the best practice document6 to national requirements • Definition of the appropriate models of care • Implementation of best practice and an evaluation of care with the ILF minimum dataset, launched in 2011

K23346_Neligan_48_How to Estab_r2_dc_0607-0616.indd 609

5/26/2015 10:14:53 AM

610

Part VI  Research and Future Directions

Establishing a National Lymphedema Framework In Lymphoedema Frameworks—The Way Forward,7 the ILF suggests that the following stages are an important process that will help ensure the success of the national organization: 1. Identify academic and clinical leads. 2. Identify stakeholders. 3. Identify the executive committee. 4. Incorporate the executive and stakeholders into a steering group. 5. Arrange steering group meetings to decide: ȤȤ Structure of the national lymphedema framework ȤȤ Roles and responsibilities ȤȤ Definition of the goals ȤȤ Scope of work ȤȤ Timelines ȤȤ Funding strategies 6. Plan a stakeholders’ meeting. This gives all the stakeholders the opportunity to contribute to the discussion and enables the identification of issues that must be addressed. In addition, this meeting helps identify possible solutions and possibly other stakeholders. 7. This meeting will culminate in a strategic plan for the national framework and identify working groups that are needed to ensure that the process is implemented. 8. The national framework will then undertake a scoping exercise that determines current practice and suggests where developments are needed so that best practice can be implemented. 9. The international Best Practice for the Management of Lymphoedema6 should be adapted if necessary to meet the needs of the country. 10. From this the following steps will occur: ȤȤ Appropriate models of care will be defined. ȤȤ Definition of a strategy that uses the ILF minimum dataset as the evaluation tool will be established. ȤȤ The working group will collaborate with reimbursement agencies to ensure that all their required information is included in this dataset. ȤȤ Best practices will be implemented within the service with the use of the minimum dataset to measure health outcomes and cost-effectiveness. The outcomes from this preparatory work will ensure that improvements in practice and reimbursements will be recognized.

Structure of a National Lymphedema Framework The overall structure of the framework may vary according to the needs of that country but usually includes an executive committee for the day-to-day management and a steering group of key stakeholders. These stakeholders must have the motivation and capacity to drive change.

K23346_Neligan_48_How to Estab_r2_dc_0607-0616.indd 610

5/26/2015 10:14:53 AM

Chapter 48  Establishment of a Lymphedema Framework

611

Within the structure a prominent academic or clinician who has standing within his or her country, the leadership skills, and a vision for the project must be involved. Also, working groups should be formed to undertake key work according to the needs of the country. The national frameworks interrelate with the ILF in several ways, including membership of the steering group, collaboration on international projects, and a leadership role on projects of particular interest. The frameworks are asked to be part of and maintain the ethos of the ILF, including the sharing of tools and expertise to help each other.

Funding of National Lymphedema Frameworks The ILF recommends that a national framework should be established as a not-for-profit organization. Funding opportunities may exist from the following sources: 1. State funding: This funding should be investigated, particularly for the research aspects of the work. It will be necessary to show prospective funders that the national lymphedema framework has the long-term capacity to define a model of care that will contribute to better patient care and help reduce the cost of treatment. However, state funding is very competitive. 2. Charitable funding: It can be time-consuming to explore prospective charitable organizations. However, some frameworks have found that charitable funding is an invaluable source of support, and thus these organizations should be actively considered. 3. Industry: Industry, which is considered a key stakeholder in a national framework, brings specific expertise to the project. Industry, along with patients and practitioners, should be active contributors to working groups and possibly serve on the steering groups in a nonvoting capacity. In this manner industry will: ȤȤ Develop a vision for lymphedema. ȤȤ Understand the needs of both patients and practitioners. ȤȤ With increased knowledge develop new and improved solutions for treatment. ȤȤ Prepare for future market developments. The ILF will enable national frameworks to draw up a sponsoring package for interested companies. To obtain industry support, companies must be convinced of the benefits of their participation in the project and the ability of the framework to deliver the project. A strategy by the framework to improve the management of lymphedema in that country will enable industry, as a key stakeholder, to understand the potential for market development and expansion that will benefit patients, practitioners, and industry itself. There are ethical rules about working effectively with industry, and the national frameworks and ILF should ensure adherence to these rules. 4. Self-funding: Eventually national frameworks should generate income from their activities, such as conferences, educational programs, and workshops, and through the sale of created resources.

K23346_Neligan_48_How to Estab_r2_dc_0607-0616.indd 611

5/26/2015 10:14:53 AM

612

Part VI  Research and Future Directions

What Is Different About a Lymphedema Framework? Several aspects differentiate a lymphedema framework from other approaches that are common with many international societies: • The philosophy of both the ILF and national frameworks is based on partnership and sharing. • A national framework recognizes that patients have a particular expertise and their value is equal to that of professionals. Patients are often the initiating force in the framework They alone know what it is like to live with lymphedema. The ILF’s objective is to ensure that the role of the “expert patient,”8 who not only understands his or her own condition but can also influence the development of lymphedema care, is advanced. In the United Kingdom, patients in the Lymphoedema Support Network are actively engaged in strategically driving projects to ensure that the national drug tariff is aware of reimbursement requirements. The international dimension is to promote and facilitate collaboration and synergy. Although projects may be developed in one country, the expectation is that they will be used throughout the ILF for the common benefit. The ILF advocates that this approach provides a unique opportunity to build a strong evidence base for lymphedema management. International projects provide national data that contribute to an international view of the problem. In this way data generated from these projects are used to support other countries in their battle for reimbursement of care. In addition, the ILF recognizes that the collection of robust epidemiologic and outcome data will make it possible to lobby for improved services, reimbursed care, and thus improved patient outcomes. This type of international approach also provides great opportunities for international clinical trials that are desperately needed in this field.

Key Aspects of ILF Work Conferences International conferences are held regularly in partnership with host frameworks. This allows both traditional conference activities and workshops, as well as networking opportunities, for the frameworks. These conferences provide countries attempting to establish a national framework the opportunity to ask the ILF any questions. During the conference, working groups and expert panels also have the opportunity to meet and work face to face. This is particularly important because of the vast differences in approaches to lymphedema care worldwide.

K23346_Neligan_48_How to Estab_r2_dc_0607-0616.indd 612

5/26/2015 10:14:53 AM

Chapter 48  Establishment of a Lymphedema Framework

613

The conference always hosts a patient conference where patients with lymphedema can hear about recent research and developments in the field, participate in planned workshops, visit the exhibition of voluntary support organizations and industry, and meet with other patients with lymphedema and chronic edema.8

Children’s Lymphedema Strategy The ILF recognizes that lymphedema in children is a rare but very important problem. In many countries there is little or no provision of care and few specialist centers. Research with parents and families in France and the United Kingdom revealed their trauma when they attempted to reach expert clinicians and receive appropriate care.9 The ILF is developing an international strategy that will include outcome measures and quality of life tools for children with the condition. Documents have been launched to recommend the types of services required, and international links with genetic teams are being established.10 In 2013 the first international children’s camp was held during the ILF conference in Montpellier, France, in which children and their families came to an educational camp. For many children this was their first encounter with other children with lymphedema. Language barriers did not inhibit the process, and children, families, and professionals gained invaluable information from this exercise. Industry has addressed the needs of children by developing new materials for compression stockings and devices in response to this initiative. Also, a children’s book11 has been published and translated into several languages.

Best Practice Document and Systematic Reviews Lymphedema is a progressive, chronic condition that affects a significant number of people and can have deleterious effects on patients’ physical and psychosocial health. Although it may be greatly ameliorated by the appropriate management, many patients receive inadequate treatment, are unaware that treatment is available, or do not know where to seek help. As part of the ILF work, systematic reviews were undertaken in 2002, which culminated in the publication of the Best Practice for the Management of Lymphoedema in 2006.6 This document has been widely used and was recently translated into Japanese. The best practice document was derived from a United Kingdom national consensus on the standards of practice for people who are at risk of or who have lymphedema.6 The consensus process was launched in 2002 and was driven by the Lymphoedema Framework with input from national patient support groups, patients with lymphedema, national professional lymphedema groups, clinical experts, and industry. Production of this document included reviews by an international panel of experts and endorsement by key national lymphedema organizations. Since then the American lymphedema framework has led an ambitious project to update the systematic reviews and has published nine documents.12-20

K23346_Neligan_48_How to Estab_r2_dc_0607-0616.indd 613

5/26/2015 10:14:54 AM

614

Part VI  Research and Future Directions

Other Documents Collaboration with the national lymphedema networks has enabled the publication of further documents that cover compression therapy and hosiery, surgical intervention, management of advanced cancer, and the care of children. Collaboration and partnership with a patient group produced a document about establishing patient support groups.21-27

Research In addition to the International LIMPRINT Study, the ILF has been awarded a grant to observe the normal practice of multilayer lymphedema bandaging. This study will take place in four countries. It will not only observe the effects of the bandage systems, but also patient quality of life over 6 months and the costs of the systems used and therapists’ time factors.

Conclusion The ILF believes it is essential to work in partnership with clinicians, academics, patients, researchers, and industry. This will enable a greater understanding of the condition of lymphedema, its prevalence, its impact on patients and national health services, and the need for evidence-based treatments. National frameworks undertake this objective in their own countries, and this may be shared through the ILF, thus building a strong research platform and evidence base. The collection of this evidence is paramount in lobbying for improved services, reimbursed care, and, above all, improved outcomes for those patients suffering from this mostly unrecognized condition.

C linical P earls • Remain focused on the vision. • Concentrate on what matters. • Understand national issues. • Ensure strong leadership. • Maintain good communication. • Ensure that the vision is supported by action. • Accept collective responsibility for finding solutions. • Encourage a motivated skill mix. • Try to avoid interprofessional conflict that prevents progress. • Avoid politics where possible.

K23346_Neligan_48_How to Estab_r2_dc_0607-0616.indd 614

5/26/2015 10:14:54 AM

Chapter 48  Establishment of a Lymphedema Framework

615

R EFERENCES 1. Moffatt C, Pinnington L. HIEC Project Evaluation Report. Facilitating development of community based lymphoedema services through clinical education. A report prepared for East Midlands Health, Innovation and Education Cluster, 2012. 2. Budge PJ, Little KM, Mues KE, et al. Impact of community-based lymphoedema management on perceived disability among patients with lymphatic filariasis in Orissa State, India. PLoS Negl Trop Dis 7:e2100, 2013. 3. Mwasa ET, Stensgaard AS, Nsakashalo-Senkwe M, et al. Mapping the geographical distribution of lymphatic filariasis in Zambia. PLoS Negl Trop Dis 8:e2714, 2014. 4. Molyneux DH, Taylor MJ. Current status and future prospects of the Global Lymphatic Filariasis Programme. Curr Opin Infect Dis 14:155-159, 2001. 5. Michael E, Malecola MN, Zervos M, et al. Global eradication of lymphatic filariasis: the value of chronic disease control on parasite elimination programmes. PLoS One 3:e2936, 2008. 6. Moffat CJ, ed. Lymphoedema Framework. Best Practice for the Management of Lymphoedema. International Consensus. London: MEP Ltd, 2006. 7. Glover D, ed. Lymphoedema Frameworks—The Way Forward. London: International Lymphoedma Framework, 2010. 8. The Expert Patient: A New Approach to Chronic Disease Management for the 21st Century. London: Department of Health, September 2001. 9. Moffatt CJ, Murray SG. The experience of children and families with lymphoedema—a journey within a journey. Int Wound J 7:14-26, 2010. 10. Glover D, ed. Care of Children with Lymphoedema. London: International Lymphoedema Framework, 2010. 11. Todd J. The Big Book of Lymphoedema. Leeds: Drewton Publishing, 2009. 12. Cormier JN, Rourke L, Crosby M, et al. The surgical treatment of lymphedema: a systematic review of the contemporary literature (2004-2010). Ann Surg Oncol 19:642-651, 2012. 13. Kwan ML, Cohn JC, Armer JM, et al. Exercise in patients with lymphedema: a systematic review of the contemporary literature. J Cancer Surviv 5:320-336, 2011. 14. Lasinski BB, McKillip Thrift K, Squire D, et al. A systematic review of the evidence for complete decongestive therapy in the treatment of lymphedema from 2004-2011. PM R 4:580-601, 2012. 15. Ridner SH, Fu MR, Wanchai A, et al. Self-management of lymphedema: a systematic review of the literature from 2004 to 2011. Nurs Res 61:291-299, 2012. 16. Fu MR, Ridner S, Hu SH, et al. Psychosocial impact of lymphedema; a systematic review of literature from 2004-2011. Psychooncology 22:1466-1484, 2012. 17. Beck M, Wanchai A, Stewart BR, et al. Palliative care for cancer-related lymphedema: a systematic review. J Palliat Med 15:821-827, 2012. 18. Feldman JL, Stout NL. Intermittent pneumatic compression therapy: a systematic review. Lymphology 45:13-25, 2012. 19. Stout NL, Weiss R, Feldman JL, et al. A systematic review of care delivery models and economic analyses in lymphedema: health policy impact (2004-2011). Lymphology 46:27-41, 2013. 20. Rodrick JR, Poage E, Wanchai A, et al. Complementary, alternative, and other noncomplete decongestive therapy treatment methods in the management of lymphedema: a systematic search and review. PM R 6:250-274; quiz 274, 2014. 21. Glover D, ed. Compression Therapy: A Position Document on Compression Bandaging. Best Practice for the Management of Lymphoedema, ed 2. London: International Lymphoedema Framework, 2012. 

K23346_Neligan_48_How to Estab_r2_dc_0607-0616.indd 615

5/26/2015 10:14:54 AM

616

Part VI  Research and Future Directions

22. Glover D, ed. Surgical Intervention: A Position Document on Surgery for Lymphoedema. Best Practice for the Management of Lymophoedema, ed 2. London: International Lymphoedema Framework, 2012. 23. Glover D, ed. International Lymphoedema Framework and Canadian Lymphedema Framework. The Management of Lymphoedema in Advanced Cancer and Oedema at the End of Life. London: International Lymphoedema Framework, 2010. 24. MacGregor L, ed. Lymphoedema Framework. Template for Practice: Compression Hosiery in Lymphoedema. London: MEP Ltd, 2006. 25. MacGregor L, ed. Lymphoedema Framework. Developing a Lymphoedema Service. London: MEP Ltd, 2007. 26. Template for Practice: Compression Hosiery in Upper Body Lymphoedema. Aberdeen: HealthComm UK Ltd, 2009. 27. Glover D, ed. International Lymphoedema Framework and Lymphoedema Support Network. Elements of an Effective National Patient Organisation: The Lymphoedema Support Network. London: International Lymphoedema Framework, 2010.

K23346_Neligan_48_How to Estab_r2_dc_0607-0616.indd 616

5/26/2015 10:14:54 AM

C redits Chapter 1 Table 1-1  From International Society of Lymphology. The diagnosis and treatment of peripheral lymphedema: 2013 Consensus Document of the International Society of Lymphology. Lymphology 46:1-11, 2013. Chapter 5 Fig. 5-5  From Taylor GI, Pan WR. The Angiosome Concept and Tissue Transfer. St Louis: Quality Medical Publishing, 2014. Figs. 5-6, 5-8, 5-10 B and C, 5-11  From Pan WR, Le Roux CM, Briggs CA. Variations in the lymphatic drainage pattern of the head and neck: further anatomic studies and clinical implications. Plast Reconstr Surg 127:611-620, 2011. Fig. 5-7  From Pan WR, Suami H, Taylor GI. The superficial lymphatic drainage of the head and neck: an anatomical study and clinical implication. Plast Reconstr Surg 121:1614-1624, 2008. Fig. 5-12  Pan WR, Le Roux CM, Levy SM, et al. Lymphatic drainage of the external ear. Head Neck 33:60-64, 2011. Fig. 5-16  From Suami H, Pan WR, Mann GB, et al. The lymphatic anatomy of the breast and its implications for sentinel lymph node biopsy: a human cadaver study. Ann Surg Oncol 15:863-871, 2008. Figs. 5-23, 5-26  From Pan WR, Wang D-G, Levy SM, et al. Superficial lymphatic drainage of the lower extremity: anatomic study and clinical implications. Plast Reconstr Surg 132:696-707, 2013. Figs. 5-30, 5-31  From Pan WR, Le Roux CM, Levy SM. Alternative lymphatic drainage routes from the lateral heel to the inguinal lymph nodes: anatomic study and clinical implications. ANZ J Surg 81:431-435, 2011. Chapter 8 Fig. 8-1  From Martinez-Corral I, Makinen T. Regulation of lymphatic vascular morphogenesis: implications for pathological (tumor) lymphangiogenesis. Exp Cell Res 319:1618-1625, 2013.

Fig. 8-2  From Tammela T, Alitalo K. Lymphangiogenesis: molecular mechanisms. Cell 140:460-476, 2010. Fig. 8-3  From Connell FC, Gordon K, Brice G, et al. The classification and diagnostic algorithm for primary lymphatic dysplasia: an update from 2010 to include molecular findings. Clin Genet 84:303-314, 2013. Chapter 10 Table 10-1  Data from Belov ST. Anatomopathological classification of congenital vascular defects. Semin Vasc Surg 6:219224, 1993; Rutherford RB. Classification of peripheral congenital vascular malformations. In Ernst C, Stanley J, eds. Current Therapy in Vascular Surgery. St Louis: Mosby, 1995; and Lee BB, Do YS, Yakes W, et al. Management of arteriovenous malformations: a multidisciplinary approach. J Vasc Surg 39:590600, 2004. Table 10-3  From HUGO Gene Nomenclature Committee. Hepatocyte growth factor (hepapoietin A; scatter factor). Available at http://www.genenames.org/cgi-bin/gene_symbol_ report?hgnc_id5HGNC:4893. Chapter 14 Fig. 14-4  A, Courtesy of Dr. B Vijaya, Department of Pathology, JSS Medical College, JSS University, Mysore, India. Chapter 22 Fig. 22-1  From The Human Protein Atlas. Available at http:// www.proteinatlas.org/; and from Uhlén M, Fagerberg L, Hallström BM, et al. Tissue-based map of the human proteome. Science 347(6220):1260419, 2015. Chapter 23 Table 23-1  From the International Society of Lymphology. The diagnosis and treatment of peripheral lymphedema: 2013 Consensus Document of the International Society of Lymphology. Lymphology 46:1-11, 2013. Table 23-2  From Földi M, Földi E, eds. Földi’s Textbook of Lymphology. San Francisco: Elsevier, 2009.

617

K23349_Nelign_98_Credits_r5_0617-0618.indd 617

5/29/2015 8:19:14 AM

618

Credits

Table 23-3  From Campisi C, Michelini S, Boccardo F, et al. Modern stadiation of lymphedema and corresponding preventive options. Eur J Lymphol 7:27, 1999. Table 23-4  From Michelini S, Campisi C, Failla A, et al. Staging of lymphedema: comparing different proposals. Eur J Lymphol 16:7-10, 2006. Table 23-5  From Book of Abstracts, Twentieth International Congress of International Society of Lymphology, I Brazilian Congress of Lymphology and I Congreso del CAPAL (Latin American Chapter of the International Society of Lymphology), 2005. Chapter 24 Fig. 24-2  Courtesy of Fabrication Enterprises, Inc. Fig. 24-3  Courtesy of Pero-System Messgeraete GmbH. Table 24-1  From Kleinhans E, Baumeister RG, Hahn D, et al. Evaluation of transport kinetics in lymphoscintigraphy: follow-up study in patients with transplanted lymphatic vessels. Eur J Nucl Med 10:349-352, 1985. Chapter 25 Fig. 25-1  From Olszewski WL, Jain P, Ambujam G, Zaleska M, et al. Topography of accumulation of stagnant lymph and tissue fluid in soft tissues of human lymphedematous lower limbs. Lymphat Res Biol 7:239-245, 2009. Figs. 25-2, 25-3 A  From Olszewski WL, Ambujam PJ, Zaleska M, et al. Where do lymph and tissue fluid accumulate in lymphedema of the lower limbs caused by obliteration of lymphatic collectors? Lymphology 42:105-111, 2009. Figs. 25-5, 25-6, 25-12  From Olszewski WL. Contractility patterns of human leg lymphatics in various stages of obstructive lymphedema. Ann N Y Acad Sci 1131:110-118, 2008. Figs. 25-7, 25-8, 25-9  From Olszewski WL. Physiology— lymph flow. In Lee BB, Bergan J, Rockson SG, eds. Lymphedema: A Concise Compendium of Theory and Practice. London: Springer, 2011. Fig. 25-17  From Olszewski WL, Cwikla J, Zaleska M, et al. Pathways of lymph and tissue fluid flow during intermittent pneumatic massage of lower limbs with obstructive lymphedema. Lymphology 44:54-64, 2011. Figs. 25-18, 25-19, 25-20, 25-21, 25-23  From Zaleska M, Ols­zewski WL, Jain P, et al. Pressures and timing of intermittent pneumatic compression devices for efficient tissue fluid and lymph flow in limbs with lymphedema. Lymphat Res Biol 11:227-232, 2013.

K23349_Nelign_98_Credits_r5_0617-0618.indd 618

Fig. 25-22  From Olszewski WL, Jain P, Ambujam G, et al. Tissue fluid pressure and flow during pneumatic compression in lymphedema of lower limbs. Lymphat Res Biol 9:77-83, 2011. Chapter 30 Box 30-1  Data from Keeley V. Drugs that may exacerbate and those used to treat lymphoedema. J Lymphoedema 3:5765, 2008. Chapter 33 Box 33-1  From Boccardo F, Casabona F, De Cian F, et al. Lymphedema microsurgical preventive healing approach: a new technique for primary prevention of arm lymphedema after mastectomy. Ann Surg Oncol 16:703-708, 2009. Chapter 35 Fig. 35-1  From Koshima I, Kawada S, Moriguchi T, et al. Ultrastructural observations of lymphatic vessels in lymphedema in human extremities. Plast Reconstr Surg 97:397-405; discussion 406-407, 1996. © Georg Thieme Verlag KG. Fig. 35-2  From Mitsushima I, Narushima M, Mihara M, et al. Lymphatic anastomosis for lymphedema. Obstet Gynecol 47:183-189, 2008. Figs. 35-4, 35-5  From Koshima I, Nanba Y, Tsutsui T, et al. Long-term follow-up after lymphaticovenular anastomosis for lymphedema in the leg. J Reconstr Microsurg 19:209-215, 2003. Figs. 35-6, 35-7  From Koshima I, Inagawa K, Urushibara K, et al. Supermicrosurgical lymphaticovenular anastomosis for the treatment of lymphedema in the upper extremities. J Reconstr Microsurg 16:437-442, 2000. © Georg Thieme Verlag KG. Chapter 37 Figs. 37-1 A, 37-2, 37-4  Produced with permission from Mount Sinai Health System. Chapter 44 Fig. 44-1  From Sabesan S, Vanamail P, Raju KH, Jambulingam P. Lymphatic filariasis in India: epidemiology and control measures. J Postgrad Med 56:232-238, 2010. Chapter 45 Fig. 45-1  Liu NF, Yan ZX, Wu XF. Classification of lymphatic-­ system malformations in primary lymphoedema based on MR lymphangiography. Eur J Vasc Endovasc Surg 44:345-349, 2012.

5/29/2015 8:19:14 AM

I ndex A Ablative surgery, 12, 13; see also Excisional surgery Acanthosis, 196 Activation, lymphatic, in immune response, 164-167 Acute lymphedema, 190 Adherence, in lymphedema management, 408, 559 Adipocyte proliferation, 4, 5 Adipogenesis, 136, 138-140 Adiponectin, 140 Adipose tissue in secondary lymphedema, 207-208, 209 Adipose tissue deposition adipogenesis and, 136 in chronic lymphedema, 437-438 effects of, 561-562 in elephantiasis, 135 lymphangiogenesis and, 136-140 Adipose-derived stem cells (ADSCs), 595, 599-601; see also Stem cells Adolescence, lymphedema in, 4; see also Pediatric patients Advanced lymphedema, multiple lymphaticovenous anastomoses in, 457-458 Aesculus hippocastanum, 410-411 Agarwal technique, 223-224 Air travel, 258 AK1 mutations, 129-130 Albendazole, 222, 571 Albumin, in technetium-99m tracers, 356 Alternative medicine; see Complementary and alternative medicine (CAM) American horse chestnut, 8 Amniotic band syndrome, 240 Angiogenesis, in embryology of vascular system, 58; see also Lymphangiogenesis Angiography; see Computed tomography (CT); Magnetic resonance lymphangiography (MRL) Animal models genetic, 590-591 list of, 584 surgical, 585-590 validity of, 583-585 Ankle, lymphatic vessels in, 85, 86 Anopheles, 569 Anterior cardinal vein, 53, 58

Anterior cervical lymphatic vessels, 63, 64, 71 Anterior group in ankle, 86 in foot, 85 in knee, 87 in thigh, 88 Anterior radial pathway, 104, 108 Anterior ulnar pathway, 104, 108 Anterolateral group in arm, 83 in leg, 87 Anteromedial group, in leg, 87 Antibiotics, for cellulitis, 414-415 Arm; see Forearm; Upper limb Arm dermal backflow (ADB) stage, 370 Arteriography, 155 Autologous lymph node transfer (ALNT); see Vascularized lymph node transfer (VLNT) Axillary blockage, lymphatic grafting for, 465-467 Axillary lymph node transfer, 491-493, 507-509, 514517 Axillary node in collecting system of breast, 77, 78 dissection, 101, 256-257 in drainage of upper limb, 105 Axillary pedicle, 102-103 Axillary-vein chain, 105 Axillary-vein pedicle, 103 Ayurvedic oil, 211 Azygos vein, in thoracic duct anatomy, 99, 100 B Bacteria, common entry points for, 204-205 Baker cyst, 292 Basilic pathway, 105, 108 Benzopyrones, 8, 408-410 Bevacizumab, 602 Bicipital pathway, 105, 108 Bilateral lower limb lymphedema, frequency and prevention of, 483-484 Bioimpedance spectroscopy (BIS), 322-323 Biomarker(s) CD31 as, 300, 301 CD44 as, 302

619

K23349_Nelign_99_Index_r3_0619-0634.indd 619

5/27/2015 2:12:45 PM

620

Index

Biomarker(s)­—cont'd ecto-5'-nucleotidase as, 300, 301 LYVE1 as, 302, 303 mannose receptor, C type 1 as, 304-305 podoplanin as, 302-304 PROX1 as, 300, 301 VEGFR-3 as, 304 Biopsy, of sentinel lymph nodes accuracy and sensitivity of, 266 in breast/anterior torso, 79 in breast cancer, 264, 268, 270-271 in head and neck, 75-76 history of, 263-264 long-term survival outcomes and, 266 in lower limb, 93-94 lymphedema outcomes and, 267-269 lymphedema risk after breast cancer with, 256 in melanoma, 265, 267 outcome impact of, 266-271 quality of life outcomes and, 269-271 surgical technique for, 265-266 Bleomycin, 157 Blue patent violet (BPV), 451 Blue rubber bleb nevus, 149, 283 Bone morphogenic protein (BMP), 596 Brachiocephalic trunk, 100 Brawny edema, 202 Brazilian Society of Phlebology and Lymphology, 312 Breast collectors in, 76-79 cutaneous lymphatic network in, 101 drainage of, 101-114 lymphatic pedicles in, 102-104 Breast cancer incidence of, 264 quality of life outcomes in, 270-271 radiotherapy in, 257 sentinel lymph node biopsy in, 264, 268, 270-271 Breast cancer–related lymphedema (BCRL) axillary node dissection in, 256-257 clinical evaluation of, 540 CT angiography in, 543-544 exercise in patients with, 257-258 imaging of, 541-544 incidence of, 4-5, 26-27 in India, 571-572 indocyanine green lymphography in, 541-542 lymph node transfer in, 545-547 lymphatic microsurgical preventing healing approach in, 527-531 lymphaticovenular anastomosis in, 544-545 lymphoscintigraphy in, 542-543 MR lymphography in, 543 obesity and, 27, 258 palliative approaches to, 539 preoperative assessment for, 540-544 reconstructive approaches to, 539-540

K23349_Nelign_99_Index_r3_0619-0634.indd 620

risk factors for lymphedema in, 256-260 surgical techniques for, 544-547 treatment algorithm for, 547, 548 Breast-conserving therapy (BCT), 256-257 Bridge precollectors, 64, 65, 66 Bromelain, 8 Broncho-mediastinal trunk left, 54 right, 54 Brucellosis, 240 Brugia malayi, 166-167, 216, 569; see also Lymphatic filariasis Brugia timori, 166-167, 216; see also Lymphatic filariasis Butcher’s broom, 411 Bypass, lymphatic-lymphatic, 13-14 C Cadaveric studies, 62 Cadherin, 124 Collagen and calcium-binding endothelial growth factor domain 1 (CCBE1) in lymphangiogenesis, 123, 124 lymphatic endothelial cells and, 597 mutations, 126, 128 Calf bandaging of, 340-341 manual massage of, 339, 340 pneumatic compression of, 342 Cancer treatment, 4-5; see also Breast cancer Cancer-specific quality of life tools, 45 Canine models, 584, 585-586 Capillaries, lymphatic, 55, 98 Capillary bed, 53-54, 196 Capillary malformations, 147-148 Cardiovascular complications, 560 Cavernous lymphangioma, 282 CD31, 300, 301 CD44, 302 Cellulitis antibiotics for, 414-415 as differential diagnosis, 292, 293 as morbidity of lymphedema, 564 tissue fluids and, 196 Cephalic derivative pathway, 106 Cephalic pathway, 105, 108 Cervical cancer, incidence of, 264-265 Charles procedure, 12, 13, 425-429, 575 Charles’s excision, 226 Chemotherapy in breast cancer, as lymphedema risk factor, 257 in lymphatic filariasis, 222 Children; see Pediatric patients China, 573-579 Chronic edema, as term, 608 Chronic obstructive pulmonary disease (COPD), lymphatic vessels in, 164

5/27/2015 2:12:45 PM

Index

Chronic venous insufficiency, 293 Chyle, 236 Chylocele, 200 Chylometrorrhagia, 200 Chylothorax, 200 Chylous ascites, 200 Chylous reflux, 200, 237, 402 Chyluria, 218 Circumflex iliac group, 109 Cirrhosis, 293 Cirsoid aneurysm, 143-144 Cisterna chyli in anatomy, 54, 100 in embryology, 59 in thoracic duct anatomy, 99 Clindamycin, 415 Clothing, 566-567 CLOVES, 126, 239 Collecting lymphatic vessels, 54-55, 56 Combined lymphedema surgical treatment (CLST), 547, 548 Combined physical therapy (CPT), 7-8 Combined vascular malformation, genetics in, 126 Common cardinal veins, 58, 145 Communicating system, 99 Comorbidities quality of life tools and, 47 in secondary lymphedema, 192 Complementary and alternative medicine (CAM) herbal pharmacotherapies, 410-412 quality of life and, 35 as treatment, 8 Complete decongestive physiotherapy (CDP), 395; see also Treatment Complete decongestive treatment (CDT), 395, 572; see also Treatment Complete lymphedema functional treatment (CLyFT), 455-456; see also Treatment(s) Compliance, patient, 408, 559 Complications of lymphedema cardiovascular, 560 examples of, 560 genitourinary, 560, 561 infection as, 563-564 integumentary, 563 malignant transformation as, 564 massive localized lymphedema as, 565 musculoskeletal, 561-562 pain as, 565 in primary versus secondary, 559-560 psychosocial, 566-567 in specific body systems, 560-563 surgical in Agarwal technique, 225 in liposuction, 442-443 in lymph node transfer, 523

K23349_Nelign_99_Index_r3_0619-0634.indd 621

621

in Manokaran technique, 226 in nodovenous shunt, 229 Comprehensive medical care, 395; see also Treatment Compression after liposuction, 442 as conservative treatment, 397 in Földi method, 476 hydrodynamics in, 337-347 intermittent pneumatic, 9, 341-346, 397 lymphatic malformations and, 157 lymphaticovenular anastomosis with versus without, 479 maintenance phase in, 397 in phlebolymphedema, 251 Computed tomography (CT) angiography, in breast cancer–related lymphedema, 543-544 in diagnosis, 295 in phlebolymphedema, 250 single photon emission, 295 of vascular malformations, 155 Congenital conditions, 281-282 Congenital unisegmental edema, genetics in, 126 Congenital vascular malformations (CVMs), lymphatic malformations and, 177; see also Vascular malformations Congestive heart failure, 293, 560 Constipation, 402 Coumarin, 398-399, 408-410, 411-412 COUP transcription factor, 123 Crohn’s disease, 281 Culex quinquefasciatus, 569 Culex spp., 216 Cultural considerations, with quality of life, 33 Cyclic chemotherapy, in lymphatic filariasis, 222 Cyclo 3 Fort, 398, 411 CYP26B, 596 Cystic angiomatosis, 282 D Daflon, 398 Debulking Agarwal technique for, 223-225 Charles’s excision for, 226 Manokaran technique for, 225-226 penile, 223 Deep inferior epigastric perforator (DIEP) flap, 477, 489, 491, 492, 519-520, 546 Deep inguinal nodes, 90 Deep popliteal nodes, 92 Deep system defined, 99 in lower limb, 106 Deep vein thrombosis, 292, 293 Deltotricipital pathway, 106 Dental caries, 221

5/27/2015 2:12:45 PM

622

Index

Depth of lymph vessels in subcutaneous tissue, in leg, 91-92 Derivative pathways, in upper limb, 106 Dermal backflow in diagnosis, 294 in indocyanine green lymphography, 369-373 in lymphoscintigraphy, 294, 353, 357, 358 Dermatolymphangioadenitis, 182, 221 Dextran, in technetium-99m tracers, 356 Diabetes mellitus type II, 192 Diagnosis, 293-295 differential, 292-293, 350 of lymphatic filariasis, 219-220 lymphoscintigraphy in, 156, 294, 349 in pediatric patients, 236-237 of phlebolymphedema, 248-250 as preoperative consideration, 448-449 in treatment selection, 396 of vascular malformations, 155-156 Diet, 10, 402 Diethylcarbamazine, 222, 414, 571 Differential diagnosis, 292-293, 350 Diffuse hemangiomatosis, 283 Digits, lymphatic vessels in, 80 Diosmin, 409, 410 Dipteryx odorata, 411-412 Distichiasis, genetics in, 126 Distribution of lymphatic system, 99, 180 Diuretics, 8, 416 Dog models, 584, 585-586 Dorsoradial group, 81-82 Dorsoulnar group, 81-82 Doxycycline, 157, 222, 414 Drainage; see Lymphatic drainage Drug therapy; see Pharmacologic treatment Duplex imaging, 248-249 Duplex ultrasonography; see Ultrasonography E Ear, external, lymphatic vessels of, 72-73 Economic impact, on quality of life, 31-32 Ecto-5'-nucleotidase, 300, 301 Egyptian column shape, 207 Elastin, 202-204 Electrostimulation, 398 Elephantiasis; see also Lymphatic filariasis; Primary lymphedema; Secondary lymphedema defined, 219 lymph node transfer in, 490 in lymphatic filariasis, 218 pathophysiology of, 200-201 in staging, 278, 292, 310, 448 Elephantiasis nostras verrucosa, 198 Elevation, 9-10, 222 Embolization, of vascular lesions, 157-158

K23349_Nelign_99_Index_r3_0619-0634.indd 622

Embryology lymphangiogenesis and, 122 of lymphatic endothelial cells, 596 lymphatic endothelial cells in, 122-123 of lymphatic malformations, 144-145 of lymphatic system, 57, 58-59, 136-137, 145, 278, 596 of vascular system, 58, 144-145 Embryonic cardinal vein, 57 Endermologie, 401 Endostatin, 117 Endothelial cells; see Lymphatic endothelial cells (LECs) End-to-end anastomosis, in nodovenous shunt, 228-229 End-to-side anastomosis, in nodovenous shunt, 229 Entry lesions, in lymphatic filariasis, 221 Epidermal hypertrophy, 196-197 Epifascial system defined, 99 in lower limb, 106 Epitrochlear nodes in upper limb drainage, 106 Erysipelas, 241 Escin, 410-411 Ethanol, 157 European Organization for Research and Treatment of Cancer Quality of Life Questionnaire-Core 30 Questions (EORTC QLQ-C30), 28, 29, 45 EuroQOL-5D, 27, 28, 43, 45 Evidence, for treatment, 396 Excisional surgery; see also Surgical management; specific procedures Charles procedure in, 12, 13, 425-429 future directions in, 432-433 history of, 423, 428 Homans-Miller procedure in, 427 Kondoleon procedure in, 426-427 liposuction as, 432-433 Macey procedure in, 427 patient selection for, 424-425 Sistrunk procedure in, 427 staged subcutaneous excision beneath skin flaps in, 429-432 studies of, 429 techniques, 425-433 Thompson procedure in, 427 Exercise in breast cancer patients, 257-258 mild, 400 moderate, 401 recommendations, 222 remedial, 400 as treatment, 9-10, 400-401 External ear lymphatic vessels, 72-73 External iliac nodes, 90, 92 Eye worm, 414 Eyelid lymphatic vessels, 63, 64, 69, 70 Eyelid lymphedema, 74-75

5/27/2015 2:12:45 PM

Index

F Facial dermal backflow (FDB) stage, 373 Facial region, 68-71 Family, 32-33 Fat deposition adipogenesis and, 136 in chronic lymphedema, 437-438 effects of, 561-562 in elephantiasis, 135 lymphangiogenesis and, 136-140 Fat grafting, stem cells in, 599-600 Fat wrapping, 438 Fatty acid binding protein (FABP4), 154 Fatty acids, in diet, 402 Femoral great saphenous current, 107 Fibroadipose hyperplasia, genetics in, 126 Fibroblast growth factors (FGFs), 115 Fibro-lipo-lympho aspiration, 458-459 Fibrotic lymphedema, drugs for, 415-416 Filarial lymphedema; see Lymphatic filariasis Filtration, 195 Flap(s) cervical, 494 deep inferior epigastric perforator, 477, 489, 491, 492, 519-520, 546 harvesting, from lower limb, 93 interposition, 227 lateral inguinal, 491-493 lateral thoracic, 493-494 staged subcutaneous excision beneath, 429-432 superficial inferior epigastric artery, 489, 492, 519520, 546, 547 thoracodorsal artery perforator, 513, 514-517 transverse myocutaneous gracilis, 519-524 Flavan, 409 Flavonoids, 408-410 Flow measurement, 319-321 Flow resistance, 330-332 FLQA-L, 46 Fluid transport, 181 Földi method, 476 Folliculitis, 211 Foot bandaging of, 339 lymphatic vessels in, 85-86 manual massage of, 339 pneumatic compression in, 342 superficial lymphatic system pathways in, 107 Footwear, 222 Force dissipation, 330-332 Forearm lymphatic vessels in, 82 pathways in, 104 Four-limb lymphedema, 130-131 FOXC2 in lymphangiogenesis, 124

K23349_Nelign_99_Index_r5_0619-0634.indd 623

623

mutations, 126, 129 in vascular malformations, 152-153 F.P. Weber syndrome, 238 Frameworks, 607-614 Free muscle flaps, 227 Free omental transfer, 230 Frontal lymphatic vessels, 63, 64, 66-68 Functional Assessment of Cancer Therapy-Breast Cancer (FACT-B), 45 Functional Assessment of Cancer Therapy-Melanoma (FACT-M), 29 Functional impairment, 31 G Gadobutrol, 388 Galeal layer, 65 Gap junction protein, gamma 2 (GJC2), 130-131, 154 GATA2 mutations, 126, 129 in vascular disorders, 154 Generalized lymphatic dysplasia (GLD), genetics in, 126 Genetic models, 590-591 Genetics in immune response activation, 166 in lymphangiogenesis, 121-122, 125-131 lymphatic endothelial cells and, 123-124 in primary lymphedema, 122, 125, 127-131, 179 of vascular receptors, 151-154 Genital dermal backflow (GDB) stage, 372 Genital lymphedema, lymphaticovenular anastomosis in, 484 Genitals in lymphatic filariasis, 218 Genitourinary complications, 560, 561 German Society of Lymphology, 311 Germinal center, 56 Gerota method, 98 Ginkgo, 411 Glomerulonephritis, 293 Gorham disease, 283 Gorham syndrome genetics in, 126 vascular malformations in, 149 Gorham-Stout-Haferkamp syndrome, 239 Grafting fat, stem cells in, 599-600 lymphatic for axillary blockage, 465-467 follow-up for, 470-471 harvesting in, 464-465 indications for, 469-470 in lower limb, 93, 468 in penis, 469 for peripheral blockages, 467, 468 for unilateral leg edema, 468

5/29/2015 1:03:12 PM

624

Index

Grafting—cont'd penile, 223 vascular, 463 vein, 460 Granulomatous intestinal lymphangitis, 281 Groin lymph node transfer, reverse lymphatic mapping in, 504-507 Grosmann-Rotter pedicle, 104 Growth factors, 113-116, 598-599; see also specific growth factors Gynecologic cancers, 264-265, 269, 271 H Hair tourniquet syndrome, 240 Hand, lymphatic vessels in, 81-82 Harvesting in lymphatic grafting, 464-465 Head and neck facial region in, 68-71 lymphatic vessel groups in, 63, 64 scalp region, 64-68 Health utilities, 27-28 Heart failure, congestive, 293, 560 Heating and bandage treatment, 574 Hemangioma, 144, 147-148 Hennekam syndrome genetics in, 126, 128, 131 vascular malformations in, 149 Hepatocyte growth factor (HGF) in lymphangiogenesis, 115-116 in vascular disorders, 154 Herbal pharmacotherapies, 410-412 Hesperidin, 410 History of lymphatic discovery, 62 patient, 556 Homans-Miller procedure, 427 Horse chestnut seed, 410-411 Hyaluronan receptor CD44, 302 Hydrodynamics in compression techniques, 337-347 in intermittent pneumatic compression, 341346 of lymph obstruction, 334 in normal tissue, 332-333 of subcutaneous tissue fluid flow, 336 Hygiene, in lymphatic filariasis, 221 Hyperplastic birthmarks, 147-148 Hypotrichosis-lymphedema-telangiectasia syndrome, 237 I Iliac sacs, 59 Imaging, 61-62; see also specific modalities in breast cancer–related lymphedema, 541-544 in China, 576

K23349_Nelign_99_Index_r3_0619-0634.indd 624

in phlebolymphedema, 248-249 of vascular malformations, 155-156, 158 Immune deficits, in lymphedema, 163-164 Immune function, peripheral lymphatics in, 163, 164170 Immune response lymphatic activation in, 164-167 lymphatic endothelial cells in, 164-166, 169 in lymphatic filariasis, 166-167 lymphatic regulation of, 167-170 in lymphedema pathogenesis, 170 peripheral lymphatics in, 163, 164-170 primary activation signals in, 164-166 secondary activation signals in, 166 Incidence, 26-27 India, 211, 215, 569-572 Indirect precollectors, 64, 65, 66 Indocyanine green lymphography (ICG) advantages of, 557 in breast cancer–related lymphedema, 541-542 dermal backflow in, 369-373 in diagnosis, 294-295 disadvantages of, 379-380 dynamic, 366, 367 equipment for, 366 evaluation of lymphatics in, 367-374 in functional assessment, 475 for intraoperative navigation, 374-375 lymph circulation in, 368-369 lymph transportation capacity in, 367-368 lymph vessels in, 367, 368 in lymphaticovenular anastomosis, 478 lymphedema management with, 375-376 procedures of, 366, 367 Infancy, lymphedema in, 4; see also Pediatric patients Infection, as morbidity of lymphedema, 563-564; see also Cellulitis Infectious diseases, 280-281 Inferior eyelid lymphatic branch, 69, 70 Inferior vena cava, 58 Inflammatory bowel disease, 402 Inflammatory response adipogenesis and, 138-139, 438 adipose hypertrophy, 438 benzopyrones and, 409-410 cellular migration and, 168 diet and, 402 in immune activation, 164-166 in lymphangiogenesis, 115, 116-117 lymphangiogenesis and, 138-139 in lymphangitis, 280-281 lymphatic activation and, 165-166 in lymphedema pathology, 4, 5 selenium and, 412-413 stem cells and, 599-601 Infraclavicular chain, 105

5/27/2015 2:12:46 PM

Index

Inguinal nodes alternative pathways to, from popliteal nodes, 89, 90 classification of, 109-110 in drainage of lower limb, 108-111 in lymphoscintigraphy, 358 radical clearance of, 92 Inner canthus lymphatic branch, 70 Insulin-like growth factors (IGFs), 116, 598 Integumentary complications, 563; see also Skin Interfascial system defined, 99 in lower limb, 106 Interferon gamma (IFN-gamma), 117 Interleukin-6 (IL-6), in lymphangiogenesis, 596 Interleukin-7 (IL-7), in lymphangiogenesis, 138 Intermittent pneumatic compression (IPC), 9, 341-346, 397 Internal Classification of Functioning, Disability and Health (ICF), 42 Internal thoracic vascular group, 79 International Classification of Functioning, Disability and Health (ICIDH), 313 International Lymphoedema Framework (ILF), 607609, 611-614 International Society of Lymphology (ISL), 6, 192, 207, 225, 240, 278, 295, 310-311, 321, 397, 540 Internodal lymphatic vessels, 63, 64, 72 Interpectoral pedicle, 104 Interstitial fluid; see also Tissue fluids accumulation of, 328-330 in bioimpedance spectroscopy, 322 in lymph formation, 236 in lymphedema pathology, 4-5, 180-181 VEGFR-3 mutations and, 127 Interstitial space, 98 Intertrigo, 205 Intervention; see Surgical management; Treatment(s) Intestinal lymphangiectasia, 283-284 Intestinal trunk, 54 Intraaxillary pathway, 106 Intralymphatic pressure, 198; see also Lymph pressures Intralymphatic pressure measurement, 331 Intraosseous lymphangioma, 239 Irritant soils, common entry points for, 204-205 Italian Society of Lymphangiology, 311 Ivermectin, 414 J Japanese school, 312 Jugular sacs, 53, 59 Jugular trunk, 54 Jugular-subclavian angle, 100 K Kasabach Merritt syndrome, 239 Keloid, 204

K23349_Nelign_99_Index_r3_0619-0634.indd 625

625

Kinesio/lymph taping, 402 Klinefelter syndrome, 282 Klippel-Trénaunay syndrome genetics in, 126 overview of, 283 vascular malformations in, 149, 178 Klippel-Trénaunay-like syndrome, genetics in, 126 Klippel-Trénaunay-Servelle syndrome, 238 Klippel-Trénaunay-Weber syndrome, 238 Knee lymphatic bypass grafting in, 467, 468 lymphatic vessels in, 85, 87-88 Kondoleon procedure, 426-427 L Landmarks in limb circumference measurement, 316 Lapine models, 584, 587-588 Laser therapy, 9, 399 Latency in secondary lymphedema, 191-192 Late-onset lower limb lymphedema, 130-131 Lateral cervical lymphatic vessels, 71-72 Lateral group, in foot, 86 Lateral mammary chain, 105 Lateral mammary pedicle, 102 Lateral saphenous group, 110 L-Dex ratio, 323 Leg; see also Calf; Knee; Thigh dermal backflow stage, 371 lymphatic grafting in, 468 lymphatic vessels in, 85, 86-87 Light therapy, 399 Limb circumference measurement, 220, 315, 316, 318-319 Limb volume measurement, 317-318 Lipedema, 207-208, 281, 293 Liposuction complications in, 442-443 controlled compression therapy after, 442 as excisional technique, 432-433 in fibro-lipo-lympho aspiration, 458-459 indications for, 439 operative technique for, 440-441 outcomes with, 442-443 in pediatric patients, 241 postoperative care for, 441-442 preoperative assessment for, 439 risks of, 11-12 Loa loa filariasis, 414 Lobule lymphatic vessels, 74 Localized lymphedema, massive, 565 Lodema, 398 Loiasis, 414 Loop diuretics, 416 Lower limb; see also Calf; Foot; Knee; Leg; Thigh collectors in, 84-94 drainage of, 106-111 frequency and prevention of bilateral lymphedema in, 483-484

5/27/2015 2:12:46 PM

626

Index

Lower limb­—cont'd late-onset lymphedema of, 130-131 lymph node transfer in, 493-494 secondary lymphedema of, 92-93 Low-level scanning, 399 Lumbar trunks, 54, 99 Lymph accumulation of, 328-330 characteristics of, 332-336 composition of, 54 extrinsic factors propelling, 332 formation, 236 intrinsic factors propelling, 332 Lymph flow measurement, 319-321 Lymph node anatomy, 56 functions, 56-57 number of, 56 Lymph node transfer; see Vascularized lymph node transfer (VLNT) Lymph pressures, 333, 334; see also Intralymphatic pressure Lymph sacs, 122, 124, 145 Lymph trunks, 99-100 Lymphangiodysplasias, 237, 238 Lymphangiogenesis adipogenesis and, 138-140 clinical implications of, 117-118, 125-127 defined, 113 developmental, 122 endostatin in, 117 fat deposition and, 136-140 fibroblast growth factors in, 115 genetics in, 121-122, 125-131 growth factors for stimulation of, 598-599 hepatocyte growth factor in, 115-116 in immune response activation, 164-165, 166 insulin-like growth factors in, 116 interferon gamma and, 117 interleukin-7 in, 138 intralymphatic pressure and, 198 macrophages in, 115 mechanisms inhibiting, 116-117 mechanisms promoting, 113-116 obesity and, 139 platelet-derived growth factors in, 116 primary lymphedema and, 125, 126, 127-131 stem cells and, 600 transforming growth factor beta 1 and, 116-117, 596, 600 tumor-induced, lessons from, 597-598 vascular endothelial growth factors in, 114-115, 118, 137-138, 139, 179-180, 597-598, 600 Lymphangiography in diagnosis, 294-295 in flow measurement, 320-321 in lymph node transfer, 495

K23349_Nelign_99_Index_r3_0619-0634.indd 626

lymphoscintigraphy versus, 352 nucleotide, 249 in phlebolymphedema, 249 Lymphangioleiomyomatosis, 284 Lymphangioma genetics in, 126 intraosseous, 239 vascular malformations and, 177 Lymphangioma circumscriptum, 200, 282 Lymphangiomatosis genetics in, 126 overview of, 281-282 presentation of, 282 Lymphangions, 181, 328, 332 Lymphangiosarcoma, 211, 212, 292, 564 Lymphangitis, 218, 280-281, 474 Lymphangitis carcinomatosa, 281 Lymphatic capillaries, 55, 98 Lymphatic drainage in breast, 101-114 immune function and, 167-168, 170 in lower limb, 106-111 in lymphostasis, 181-182 in secondary lymphedema, 189-190 in upper limb, 104-106 Lymphatic drainage massage, 399-400 Lymphatic duct, right, 54 Lymphatic endothelial cells (LECs) biomarkers of, 299-305 CD31 as marker for, 300, 301 cellular origin of, 596-597 ecto-5'-nucleotidase as marker for, 300, 301 in embryology of lymphatic system, 58 in immune response, 164-166, 169 in lymphangiogenesis, 113 in lymphatic capillaries, 55 LYVE1 as marker for, 302-304, 303 mannose receptor, C type 1 as marker for, 304-305 origin of, 122-123 PECAM1 as marker for, 300 podoplanin as marker for, 302-304 progenitors, 57, 59 PROX1 as marker for, 300, 301 transcription factors and, 123 Lymphatic filariasis, 4 acute attacks in, 222 Agarwal technique in, 223-224 assessment of, 220-221 atypical, 218 in China, 573-574 diagnosis of, 219-220 direct excision, 223-227 entry lesions in, 221 fat deposition in, 135 flap interposition in, 227 free muscle flaps in, 227

5/27/2015 2:12:46 PM

Index

free omental transfer in, 230 genital manifestations of, 218, 219 hygiene in, 221 in India, 569-571 lymph node transfer in, 227 lymphatic immune activation in, 166-167 lymphatic-lymphatic anastomosis in, 230 lymphaticovenous bypass in, 227 lymphosuction in, 227 manifestations of, 218-219 Manokaran technique in, 225-226 medical management of, 221-223, 413-414, 571 microfilaremia in, 217 mosquitoes in, 216 myocutaneous flaps in, 227 nodovenous shunt in, 228-229 pathogenesis of, 216-217 pharmacologic treatment of, 413-414 presentations of, 280 skin in, 5 stages of, 218 surgical management of, 223-230 worm nests in, 217 Lymphatic grafting for axillary blockage, 465-467 for axillary blockage bypass, 465-467 follow-up for, 470-471 follow-up in, 470-471 harvesting in, 464-465 indications for, 469-470 in lower limb, 93, 468 in penis, 469 for peripheral blockages, 467, 468 for unilateral leg edema, 468 Lymphatic malformations; see also Vascular malformations classification of, 144, 147 embryology of, 144-145 genetics in, 151-154 history of, 143-144 lymphangiomas and, 177 naming of, 143 tissutal, 148 truncular, 179 Lymphatic microsurgical preventing healing approach (LYMPHA), 456, 457, 554 in breast cancer, 527-530 indications for, 528, 529 long-term outcomes with, 530 in melanoma, 533, 534-535 obesity and, 528 Lymphatic obstruction deep lymphatic obstruction in, 180 extracellular volume in, 327 hydrodynamics of, 334 in lymphatic filariasis, 280 in protein-losing enteropathy, 283

K23349_Nelign_99_Index_r4_0619-0634.indd 627

627

Lymphatic pedicles, in breast, 102-104 Lymphatic system applied anatomy, 101-111 distribution of, 99, 180 embryology, 57, 58-59, 136-137, 145, 278 history of knowledge on, 62 overview, 53-57, 98 Lymphatic vessel(s) fate of, in lymphedema, 474-475 groups in breast and anterior torso, 76-79 in head and neck, 63 in upper limb, 79-84 in indocyanine green lymphography, 367, 368 Lymphatic vessel endothelial hyaluronan receptor (LYVE1), 58, 113, 122 Lymphatic-lymphatic anastomosis, 230 Lymphatic-lymphatic bypass, 13-14 Lymphaticovenous anastomoses, 14, 15 in China, 574 derivative approaches in, 450-451 dye in, 451 history of, 449-450 in lower limb, 93 multiple in advanced lymphedema, 457-458 clinical experience with, 452-454 in combined treatment protocol, 455-456 diagnostic considerations in, 448-449 dye in, 451 fibro-lipo-lympho aspiration with, 458-459 lymphoscintigraphy in, 448-449, 452-453, 460 patient selection for, 455 surgical technique for, 451-452 timing of surgery in, 456 occipital, 68 Lymphaticovenous bypass, 227, 385 Lymphaticovenous shunt, occipital, 68 Lymphaticovenular anastomosis advent of, 473 in breast cancer–related lymphedema, 544-545 compression therapy versus, 479 contraindications to, 483 at early stage, 479-480 in genital lymphedema, 484 history of, 477-478 indocyanine green lymphography in, 374-375 lateral inguinal flap in, 491-493 in long-duration lymphedema, 480-483 prophylactic, 554 recurrences in, 483 stocking use and, 476 surgical technique in, 478-479 treatment methods impact on, 476, 477 without postoperative compression therapy, 480 Lymphatic-venous-lymphatic anastomoses, 460

5/28/2015 1:06:10 PM

628

Index

Lymphedema; see also Primary lymphedema; Secondary lymphedema chronic edema versus, as term, 608 as clinical problem, 4-6 defined, 291 diagnosis of, 293-295 as immune disorder, 163-164, 170 incidence of, 26-27 nonsurgical management of, 6-11 presentation of, 291-292 staging, 6, 190-191, 292, 309-313 Lymphedema Functioning, Disability and Health Questionnaire (Lymph-ICF), 46 Lymphedema Functioning, Disability and Health Questionnaire for Lower Limb Lymphedema (Lymph-ICF-LL), 46 Lymphedema praecox, 4, 279 Lymphedema Quality of Life (LYMQOL), 28, 46 Lymphedema Quality of Life Inventory (LQOLI), 46 Lymphedema tarda, 4 Lymphedema-distichiasis syndrome, genetics in, 126, 129 Lymphnododysplasia, 178 Lymphography; see Indocyanine green lymphography (ICG); Magnetic resonance lymphography Lymphoma, 211 Lymphoscintigraphy (LSG) advantages of, 349 in breast cancer–related lymphedema, 542-543 defined, 352-356 dermal backflow in, 357, 358 in diagnosis, 156, 294, 349 in differential diagnosis, 350 in flow measurement, 319 as functional study, 352, 475 injection technique in, 357 interpretation of, 357-360 in liposuction assessment, 439 lymphangiography versus, 352 in lymphatic microsurgical preventing healing approach, 530 MRI with, 354, 356 in multiple lymphaticovenous anastomoses, 448449, 452-453, 460 normal, 358, 359 other imaging modalities versus, 349-350 in phlebolymphedema, 355 in primary lymphedema, 350, 351, 360 protocols, 357 qualitative analysis of, 353 recommendation on, 361 safety of, 354 in secondary lymphedema, 360, 361 in sentinel node mapping and biopsy, 75 in staging, 354 superficial popliteal nodes in, 111 technical aspects of, 356-357 tracers in, 356

K23349_Nelign_99_Index_r3_0619-0634.indd 628

transport index score in, 352 in treatment assessment, 354, 355 Lymphostasis, 181-182 Lymphosuction, 227 LYVE1, 302, 303, 596 M Macey procedure, 427 Macrophages, in lymphangiogenesis, 115 Maffucci syndrome malignancy in, 282-283 vascular malformations in, 149 Magnetic resonance imaging (MRI) lymphoscintigraphy with, 354, 356 of vascular malformations, 155, 158 Magnetic resonance lymphangiography (MRL) advantages of, 380, 556 in breast cancer–related lymphedema, 543 in China, 576 contrast in, 380, 382-383, 388-389 in flow measurement, 320-321 future directions in, 388-390 imaging in, 380-383 interpretation of, 383-388 in lymph node transfer, 495 magnetic resonance venography with, 381-382 patient positioning in, 381 postprocessing in, 383-388 Magnetic resonance venography (MRV) with magnetic resonance lymphangiography, 381382 in phlebolymphedema, 250 Malignant transformation, 564 Mammary gland, lymphatic drainage of, 101-102 Mannose receptor, C type 1 (MRC1), 304-305 Manokaran technique, 225-226 Manual lymphatic drainage (MLD), 7, 396-397, 408 Mapping, of sentinel lymph nodes; see also Reverse lymphatic mapping in breast/anterior torso, 79 in head and neck, 75-76 in lower limb, 93-94 Marginal sinus, 56 Massage; see also Manual lymphatic drainage (MLD) in Földi method, 476 as treatment, 399-400 Massive localized lymphedema, 565 Mastectomy, lymph node transfer with, 514-516 Measure Yourself Medical Outcome Profile (MYMOP), 42 Measurement in assessment, 220 bioimpedance spectroscopy in, 322-323 on intralymphatic pressure, 331 of limb circumference, 220, 315, 316, 318-319 of limb volume, 317-318 of lymph flow, 319-321

5/27/2015 2:12:46 PM

Index

lymphangiography in, 320-321 nuances of, 318-319 tonometry in, 321-322 Medial group in foot, 85 in knee, 87 in thigh, 88-89 in upper arm, 83 Medial saphenous group, 110 Mediastinal pedicle, 103 Medical management, of lymphatic filariasis, 221-223 Medical Outcome Survey Short-Form 36 (SF-36), 28, 29, 44 Medications; see Pharmacologic treatment Medullary sinus, 56 Meige syndrome, genetics in, 126 Melanoma, 265, 267, 270, 532, 534-535 Melilotus officinalis, 411-412 Mental lymphatic vessels, 63, 64 Mesenchymal epithelial transition factor (MET), 154 Mesenchymal stem cells (MSCs), 595, 599-600; see also Stem cells Metamerization, 147 Microcephaly with or without chorioretinopathy, lymphedema, and intellectual disability, 130 Microfilaremia, 217 Microfilariae, 413 Microsurgery, 13-17, 449-452; see also Lymphaticovenous anastomoses; Supermicrosurgery; Surgical management Midline lymphedema, quality of life tools and, 47 Migration, cellular, peripheral lymphatics in, 168-169 Milroy disease, 237 defined, 4 genetics in, 126, 131 Minocycline, 415 Models; see Animal models Monoclonal antibodies, 602 Monocyte chemotactic protein 1 (MCP-1), 596 Mosquitoes, 216 Mossy foot, 199 Mouse models, 584, 589-590 Multiple lymphaticovenous anastomoses (MLVA); see also Lymphaticovenous anastomoses in advanced lymphedema, 457-458 clinical experience with, 452-454 in combined treatment protocol, 455-456 diagnostic considerations in, 448-449 dye in, 451 fibro-lipo-lympho aspiration with, 458-459 lymphoscintigraphy in, 448-449, 452-453, 460 patient selection for, 455 surgical technique for, 451-452 timing of surgery in, 456 Murine models, 584, 588-590 Muscle cells; see Smooth muscle cells (SMCs) Musculoskeletal complications, 561-562 Myocutaneous flaps, 227; see also Flap(s)

K23349_Nelign_99_Index_r3_0619-0634.indd 629

629

N Nail changes, 206-207 Nasal lymphatic vessels, 63, 64, 69, 70 National lymphedema framework, 609-612 Natural supplements, 8 Neck; see Head and neck Neoruscogenin, 411 Neurofibromatosis type 1, 239 Neurofibromatosis type 2, 239 Neuropilin 2 (Nrp2), 123, 124, 596 Nevus flammeus, 147-148 Nipple-areola complex, 76 Nodovenous shunt, 228-229, 450 Non-Hodgkin lymphoma, 211 Nonsurgical management, 6-11 Noonan syndrome genetics in, 130 vascular malformations in, 149 Notch, 596 Notch signaling, 123, 124 Nottingham Health Profile Part 1, 45 Nucleotide lymphangiography, 249 O Obesity breast cancer–related lymphedema and, 27, 258 LYMPHA technique and, 528 lymphangiogenesis and, 139 massive localized lymphedema and, 565 weight loss for, 9 Occipital lymphatic vessels, 63, 64, 67, 68 OK-432, 157 Omega-3 fatty acids, 402 Omega-6 fatty acids, 402 Omental transfer, 230 Onychogryphosis, 206 Oral lymphatic vessels, 63, 64, 69, 70 Outer canthus lymphatic branch, 70 Ovine models, 584, 586-587 Oxerutin, 409 P Pain, 565 Parietal lymphatic vessels, 63, 64, 67, 68 Parkes-Weber syndrome genetics in, 126 vascular malformations in, 150 Passive movement, as treatment, 400 Patient compliance, 408, 559 Patient history, 556 Patient selection for excisional surgery, 424-425 for lymph node transfer, 490 for multiple lymphaticovenous anastomoses, 455 Peau d’orange, 196, 197 Pectoroaxillary pedicle, 104

5/27/2015 2:12:46 PM

630

Index

Pediatric patients children’s lymphedema strategy development for, 613 diagnosis of lymphedema in, 236-237 primary lymphedema in, 235-236, 237-239 secondary lymphedema in, 240 social aspects with, 241, 567 surgery in, 242 treatment of lymphedema in, 240-241 Penile debulking and grafting, 223, 469 Penis lymphaticovenular anastomosis in, 484 sclerosing lymphangitis of, 281 Pentoxifylline, 415 Perforating system, 99 Peripheral lymphatics in cellular migration and activation, 168-169 dysfunction, effects of, 163 in immune system, 163, 164-170 in lymphatic filariasis, 166-167 Perometer, 317-318 Pharmacologic treatment benzopyrones in, 408-410 in cellulitis, 415-416 contraindicated in lymphedema, for other conditions, 416-417 in fibrotic lymphedema, 415-416 herbal, 410-412 in lymphatic filariasis, 403, 413-414, 571 in treatment, 8-9, 398 vitamins in, 412-413 Phlebolymphedema compression in, 251 computed tomography in, 250 defined, 236, 247 diagnosis of, 248-250 imaging of, 248-250 lymphoscintigraphy in, 355 magnetic resonance venography in, 250 mechanism of, 248 as secondary lymphedema, 240 treatment of, 250-251 Physical therapy, 7-8, 490 Pig models, 584, 587 Placebo, 403 Placental growth factor (PlGF), 137 Plantar lymphatic vessels, 107 Platelet endothelial cell adhesion molecule 1 (PECAM1), 300 Platelet-derived growth factors (PDGFs), 116 Plethysmography, 248, 317 Pneumomassage, 9, 341-346; see also Compression Podoconiosis, 199, 204, 240 Podoplanin, 113, 302-304, 596 Polycystic ovarian syndrome (PCOS), 192 Popliteal fossa, 90

K23349_Nelign_99_Index_r3_0619-0634.indd 630

Popliteal nodes alternative pathways from, to inguinal nodes, 89, 90 superficial, 110 Porcine models, 584, 587 Porot test, 241 Port-wine stain, 147-148 Posterior cardinal veins, 58 Posterior group in ankle, 86 in knee, 88 in leg, 87 in thigh, 89 Posterior pathway, in arm, 105, 108 Posterior radial pathway, 104, 108 Posterior ulnar pathway, 104, 108 Posterolateral group, 83 Preauricular node, 70 Precollecting vessels, 64, 65, 66 Prepectoral pedicle, 103 Pressure; see Intralymphatic pressure; Lymph pressures Primary follicle, 56 Primary lymphedema causes of, 237 classification of, 279 complications in, versus secondary, 559-560 genetics in, 122, 125, 127-131, 179 lymphangiogenesis and, 125, 126, 127-131 lymphatic malformations in, 61, 148, 178 lymphoscintigraphy in, 350, 351, 360 lymphostasis and, 181-182 overview of, 4, 121 in pediatric patients, 237-239 survival rates in, 235 syndromes associated with, 238-239 ultrasonography in, 355-356 vascular endothelial growth factors in, 114 Primary valves, 124 Proanthocyanidins, 409 Prophylactic lymphaticovenular anastomosis, 554 Prospero homeobox 1 (Prox1), 113, 123, 124 Protein-losing enteropathy, 283-284 Proteus syndrome, 239, 283 genetics in, 126, 129 vascular malformations in, 150 PROX1, 300, 301 Psychological distress, 31, 566-567 Psychosocial Adjustment to Illness Scale (PAIS), 28, 29 Psychosocial outcomes, 30 Pudendal group, 109 “Puffy hand,” 240 Q Quality of life (QOL) assessment of, 27-30, 250-251 in breast cancer, 270-271 cultural considerations with, 33

5/27/2015 2:12:46 PM

Index

defined, 41-43 economic impact and, 31-32 family and, 32-33 functional impairment and, 31 in genital lymphedema, 484 in gynecologic cancers, 271 instruments, 28-30, 42-48 cancer-specific, 45 comorbidities and, 47 floor and ceiling effects with, 47 frequency of use of, 48 language translations with, 47 length of, 48 in lymphedema studies, 44-46 lymphedema-specific, 45-46 midline lymphedema and, 47 minimum clinically important differences in, 47 in research, 43 use of, 43-44 validation process with, 44 interventions to enhance, 34-35 psychological distress and, 31 psychosocial outcomes and, 30 rehabilitation and, 34-35 sentinel lymph node biopsy and, 269-271 spirituality and, 33 with vascular malformations, 158 Quality-adjusted life year (QALY), 43-44 Quercetin, 409 R Rabbit models, 587-588 Radial group, 82 Radionuclide lymphoscintigraphy; see Lymphoscintigraphy (LSG) Radiopharmaceuticals, in lymphoscintigraphy, 357 Radiotherapy in breast cancer, as lymphedema risk factor, 257 interventional timing and, 557 for vascular malformations, 157 Rat models, 584, 588-589 Rehabilitation, quality of life and, 34-35 Religion, 33 Rendu-Osler-Weber syndrome, vascular malformations in, 150 Retropectoral pedicle, 104 Reverse lymphatic mapping anatomy in, 504-505, 507-508 in breast cancer, 527-538 concept of, 504 in groin, 504-507 in supraclavicular region, 510 surgical technique in, 505-507, 508-509 Rhytidectomy, 74-75 Right lymphatic duct, 54 Riley-Smith syndrome, vascular malformations in, 150

K23349_Nelign_99_Index_r3_0619-0634.indd 631

631

River blindness, 413 Robles’ disease, 413 Ruscogenin, 411 Ruscus aculeatus, 411 Rutin, 409 Rutosides, 409 S Safety, of lymphoscintigraphy, 354 Scalp, 64-68 Schedule for Evaluation of Individual Quality of Life (SEIQOL-DW), 42 Sclerosing, of vascular malformations, 157, 158 Sclerosing lymphangitis of penis, 281 Scrotoplasty, 223 Secondary follicle, 56 Secondary lymphedema; see also Breast cancer–related lymphedema (BCRL) acute, 190 adipose tissue in, 207-208, 209 breast cancer surgery and, 84 causes of, 279-280 comorbidities in, 192 complications in, versus primary, 559-560 elastin in, 202-204 entry points for bacteria and irritant soils in, 204205 hepatocyte growth factor in, 116 increase in, 148 latency in, 191-192 in lower limb, 92 lymphatic drainage and, 189-190 lymphatic grafting for, 93 lymphaticovenous anastomoses for, 93 lymphoscintigraphy in, 360, 361 nail changes in, 206-207 overview of, 4, 189 pathomorphology of, 190-192 pathophysiology of, 190-192 in pediatric patients, 240 staging of, 190-191 survival rates in, 235 tissue hypertrophy in, 200-202 trauma in, 26 in upper limb, 84 vascular endothelial growth factors in, 114 venous reflux in, 209-210 Selenium, 8, 412-413 Sentinel lymph node, defined, 101 Sentinel lymph node biopsy (SLNB) accuracy and sensitivity of, 266 in breast/anterior torso, 79 in breast cancer, 264, 268, 270-271 in head and neck, 75-76 history of, 263-264 long-term survival outcomes and, 266

5/27/2015 2:12:47 PM

632

Index

Sentinel lymph node biopsy (SLNB)—cont'd in lower limb, 93-94 lymphedema outcomes and, 267-269 lymphedema risk after breast cancer with, 256 in melanoma, 265, 267 outcome impact of, 266-271 quality of life outcomes and, 269-271 surgical technique for, 265-266 Servelle-Martorell syndrome, vascular malformations in, 150 Sheep models, 584, 586-587 Shoes, 222 Short Form (36) (SF-36), 28, 29, 44 Single photon emission computed tomography (SPECT), 295 Sistrunk procedure, 427 65% point, 220 Ski jump nail, 206 Skin common entry points for bacteria and irritant soils in, 204-205 dermatolymphangioadenitis in, 182 fluid distribution in, 330 infections, 563-564 in lipedema, 207-208 lymphedema complications in, 563 venous reflux and, 209-210 water content of, 196 Skin care, 10-11 Smooth muscle cells (SMCs) after lymph node resection, 474 in lymphatic remodeling, 124, 125 regeneration of, 475 Sodium tetradecyl sulfate, 157 Soils, irritant, common entry points for, 204-205 Sonography, 220-221 South American school, 312 Sox18, 237 in lymphangiogenesis, 123 mutations, 128 in vascular malformations, 153 Spirituality, 33 Sprouting, 124 Staged subcutaneous excision beneath skin flaps, 429432 Staging, 309-313 Brazilian Society of Phlebology and Lymphology, 312 Campisi, 448, 452 dermal backflow, 369-374 in differential diagnosis, 292 German Society of Lymphology, 311 indocyanine green lymphography in, 366, 367 ISL, 6, 310-311 Italian Society of Lymphangiology, 311 Japanese, 312 lymphoscintigraphy in, 351, 354

K23349_Nelign_99_Index_r3_0619-0634.indd 632

of secondary lymphedema, 190-191 treatment decisions and, 554 Staining techniques, 98 Starling equation, 4 Starling forces, 4 Starling’s law, 196 Stem cells in adipocyte formation, 136 adipose-derived, 595, 599-601 lymphangiogenic properties of, 600 mesenchymal, 595, 599-600 as treatment, 601-603 in China, 575 in fat grafting, 599-600 sources of, 595 Stemmer sign, 202, 293 Stewart-Treves syndrome, 292, 293, 564 Stockings, 7, 9, 476, 477, 613 String sign, 218 Sturge-Weber syndrome, vascular malformations in, 150 Subaponeurotic system, 99 Subareola group, 78 Subareolar plexus, 102 Subclavian trunk, 54 Subclavian vein left, 54 right, 54-55 Subcutaneous tissue fluid pressures, 335-336; see also Tissue fluid pressure Subfascial system, 99 Submandibular node, 70 Subpectoral pedicle, 103 Subscapular chain, 105 Subscapular pedicle, 103 Sulfur, in technetium-99m tracer, 356 Superficial epigastric group, 109 Superficial inferior epigastric artery (SIEA) flap, 489, 492, 519-520, 546, 547 Superficial inguinal nodes, 90 Superficial popliteal node, 90, 91, 92, 110 Superficial system defined, 99 in lower limb, 106, 107-108 Superficial venous pump, 107 Superior thoracic pedicle, 103 Superior vena cava, 58, 99, 100 Supermicrosurgery, 473, 477; see also Lymphaticovenular anastomosis Supraclavicular lymphatic vessels, 64, 72 Supraclavicular nodes, 72 Supraclavicular reverse lymphatic mapping, 510 Supratrapezoid lymphatic vessels, 64, 72 Surgical management, 11-17; see also specific procedures ablative, 12, 13 in China, 574-576

5/27/2015 2:12:47 PM

Index

excisional Charles procedure in, 12, 13, 425-429 future directions in, 432-433 history of, 423, 428 Homans-Miller procedure in, 427 Kondoleon procedure in, 426-427 liposuction as, 432-433 Macey procedure in, 427 patient selection for, 424-425 Sistrunk procedure in, 427 staged subcutaneous excision beneath skin flaps in, 429-432 studies of, 429 techniques, 425-433 Thompson procedure in, 427 of lymphatic filariasis, 223-230 microsurgery in, 13-17, 449-452 in pediatric patients, 242 of vascular malformations, 158 Survival rates, 235, 266 Sweet clover, 411-412 T Taping, 402 Technetium-99m albumin colloid, 356 Technetium-99m dextran, 356 Technetium-99m human serum albumin, 356 Technetium-99m phytate, 356 Technetium-99m sulfur colloid, 356 Thermal therapy, 8 Thigh lymphatic vessels in, 88-91 pneumatic compression of, 342 ventromedial bundle in, as harvesting site, 464-465 Thompson procedure, 427 Thoracic duct in anatomy, 54 cervical portion of, 99 in embryology, 59 Thoracic inlet, in anatomy, 100 Thoracodorsal artery perforator (TDAP) flap, 513, 514517 Tibial great saphenous current, 107 Tissue engineering, 599 Tissue fluid pressure in lymphedema, 335-336 measurement of, 332 normal, 335 in pneumatic compression, 342-346 Tissue fluids; see also Interstitial fluid accumulation of, 328-330 characteristics of, 332-336 normal and pathologic, 196-199 Tissue hypertrophy, 200-202 Tissue manipulation, 401 Tocopherol acetate, 415

K23349_Nelign_99_Index_r3_0619-0634.indd 633

633

Toes intertrigo between, 205 lymphatic vessels in, 84 Toll-like receptors (TLRs), in immune response, 164165 Tonka beans, 411-412 Tonometry, 321-322 Torso, anterior, as lymphatic region, 76-79 Total superficial lymphangiectomy, 12 Trabecula, 56 Tracers in lymphography, 366 in lymphoscintigraphy, 356 Transcription factors, 123 Transforming growth factor beta 1 (TGF-beta 1) in lymphangiogenesis, 116-117, 123, 139, 596, 600 in lymphatic filariasis, 167 Transpectoral pedicle, 104 Transport index (TI) score, 319-320, 352 Transverse myocutaneous gracilis (TMG) flap, 519-524 Trauma-induced lymphedemas, 240 Treatment(s); see also Pharmacologic treatment; Surgical management; specific treatments adherence, 408, 559 complementary and alternative medicine as, 8, 410412 contemporary conservative, 397-402 diagnosis and, 396 evidence for, 396 goals of, 395 in India, 569-572 lymphaticovenular anastomoses and, 476, 477 lymphoscintigraphy in assessment of, 354, 355 in pediatric patients, 240-241 of phlebolymphedema, 250-251 placebo effect and, 403 prophylactic, 554 staging and, 554 traditional conservative, 396-397 of vascular malformations, 156-158 Troxerutin, 409 Truncular lymphatic malformation, 179; see also Lymphatic malformations Tuberculosis, 240 Turner syndrome, 130, 282 Type 2 diabetes, 192 U Ulnar group, 82 Ultrasonography in phlebolymphedema, 250 in primary lymphedema, 355-356 of vascular malformations, 155 Upper arm axillary blockage bypass in, 465-467 lymphatic vessels in, 83

5/27/2015 2:12:47 PM

634

Index

Upper limb axillary node dissection and lymphedema of, 101 collectors in, 79-84 drainage of, 104-106 lymph node transfer in, 491-493, 507-509, 514-517 Upper limb lymphedema 27 (ULL-27), 28, 29, 46 Upper thoracic chain, 105 V Vagus nerve, 100 Valves, intraluminal, 124-125 Vancomycin, 415 Vascular cell adhesion molecule (VCAM-1), 154 Vascular embryology, 58, 144-145 Vascular endothelial cadherin, 124 Vascular endothelial growth factor receptor (VEGFR), as biomarker, 304 Vascular endothelial growth factors (VEGFs) 3 mutations, 127 in vascular malformations, 151, 152 C in lymphangiogenesis, 597-598 mutations, 127-128 in vascular malformations, 151 D in lymphangiogenesis, 597-598 in vascular malformations, 152 in development, 123 in immune response, 166 in lymphangiogenesis, 114-115, 137-138, 139, 179-180, 597-598, 600 in lymphatic endothelial cell sprouting, 124 in lymphatic remodeling, 596-597 in oncology, 118 Vascular malformations; see also Lymphatic malformations arterial, 146, 147 arteriovenous, 146-147 classification of, 146-150 complex, 282-283 diagnosis of, 155-156 genetics in, 151-154 hemangiomas and, 144 imaging of, 155, 158 lymphangioma and, 177 quality of life with, 158 treatment of, 156-158 vascular endothelial growth factors in, 151-152 venous, 146, 147 Vascularized lymph node transfer (VLNT) in breast cancer–related lymphedema, 545-547 breast reconstruction with, 516, 517 cervical flap in, 494 complications in, 523 for congenital lower limb edema, 495

K23349_Nelign_99_Index_r3_0619-0634.indd 634

difficulty of, 488 donor sites in, 489 indications for, 488-489 lateral thoracic flap in, 493-494 local flaps and, 513-517 location choice for, 501, 555 in lower extremity, 493-494 with mastectomy, 514-516 outcomes in, 489-490, 523-524 overview of, 15-16, 227 patient selection for, 490 physical therapy with, 490 postoperative care for, 523 results with, 495-500 reverse lymphatic mapping in, 503-510 scar tissue excision and, 488 surgical technique for, 491-500 transverse myocutaneous gracilis with, 519-524 in upper extremity, 491-493, 507-509, 514-517 Vasculitis, 192 Vasculogenesis, 58 Vein graft, 460 Venography, 155, 250, 381-382 Venolymphatic malformations, 157; see also Lymphatic malformations; Vascular malformations Venous plethysmography, 248 Venous reflux, 209-210 Venous sprouting, 124 Ventromedial bundle, in thigh, 464-465 Visceral system, 99 Vitamin E, 415 Vitamin therapy, 412-413 Vitelline arteries, 58 Volar group, 82 von Hippel-Lindau disease, vascular malformations in, 150 Von Recklinghausen syndrome, 239 Vulvar carcinoma, 264 W Water content, of skin, 196 Water-displacement plethysmography, 317 Weight loss, 9 Weightlifting, 401 Wesley Clinic Lymphedema Scale (WCLS), 46 WILD syndrome, genetics in, 126 Wolbachia, 222 Worm nests, 217 Wuchereria bancrofti, 4, 166-167, 216, 240, 569; see also Lymphatic filariasis Y Yellow nail syndrome, 206 Young-Laplace equation, 157

5/27/2015 2:12:47 PM