Pediatric Sedation Outside of the Operating Room: A Multispecialty International Collaboration [3 ed.] 9783030584061, 3030584062

333 15 32MB

English Pages [898] Year 2020

Report DMCA / Copyright

DOWNLOAD FILE

Polecaj historie

Pediatric Sedation Outside of the Operating Room: A Multispecialty International Collaboration [3 ed.]
 9783030584061, 3030584062

Table of contents :
Preface
Acknowledgments
Contents
Contributors
Part I: Pediatric Sedation Outside the Operating Room
1: The History of Sedation
Introduction
Inebriation, Intoxication, Hallucination, and Anesthesia
A Forme Fruste of the Sedation Continuum
Ancient History
Chinese Drug Lore
Hindu Drugs
Sumerian Drugs
Jewish Medicine
Egyptian Medicine
Classical History
Greek Medicine
Roman Medicine
Islamic Medicine
Medieval Medicine
Ether
Varied Preparations of Varying Potencies
The Scientific or Modern Epoch
The Modern Story of Anesthesia
“Modern” Sedation and Analgesia Services
The Future of Sedation
References
2: Sedation Policies, Recommendations, and Guidelines Across the Specialties and Continents
Introduction
American Academy of Pediatrics (AAP) Guidelines
American Society of Anesthesiologists (ASA) Policies and Recommendations
Center for Medicare and Medicaid Services (CMS)
The Joint Commission
American College of Emergency Physicians (ACEP) Guidelines
International Committee for the Advancement of Procedural Sedation Consensus Statement on Fasting for Procedural Sedation
American Dental Association Sedation Guidelines
British National Health Service Dental Sedation Standards
Intercollegiate Advisory Committee for Sedation in Dentistry
American Society of Gastroenterologists
International Guidelines
Summary
References
3: Procedural Sedation: Let’s Review the Basics
Introduction
Questions to Be Asked
Definitions and Concepts
Rescue
Setting
Personnel
Equipment
Monitoring
Protocols
Patients
Procedures
Other Considerations
Future Directions
Conclusion
Case Studies
References
4: Pre-sedation Assessment
Introduction
Pre-sedation Screening
History
Physical Examination
When Not to Proceed
When Referral to General Anesthesia Is Required
Fasting Guidelines and Sedation
Preparation for and Considerations in Special Populations
Asthma and Reactive Airway Disease
Autism, Developmental Delay, and Intellectual Disability
Bronchopulmonary Dysplasia
Cannabis Use
Cerebral Palsy
Congenital Heart Disease
Cystic Fibrosis
Diabetes Mellitus
Endocrinopathies
Mitochondrial Disease
Mucopolysaccharidosis
Multiple Allergies
Muscular Dystrophies
Musculoskeletal Disorders
Obesity
Obstructive Sleep Apnea
Pregnancy
Premature Infant
Psychiatric and Behavioral Disorders
Sickle Cell Disease
Syndromes
Trauma
Tuberous Sclerosis
Upper Respiratory Tract Infection
Summary
Case Studies in Pre-sedation Assessment
References
5: Sedation Scales and Discharge Criteria: How Do They Differ? Which One to Choose? Do They Really Apply to Sedation?
Introduction
Sedation Scales
The Ramsay Scale
The Observer’s Assessment of Alertness/Sedation Scale and Modified Observer’s Assessment of Alertness/Sedation Scale
The COMFORT Scale
The University of Michigan Sedation Scale
Dartmouth Operative Conditions Scale
The Sedation-Agitation Scale
Modified Aldrete Score as a Sedation Scale
Processed EEG Monitors: The Bispectral Index and Amplitude-Integrated EEG
Auditory Evoked Potentials
Other Sedation Scales
Objective, Physiologically Based Sedation Scales
Recovery and Discharge Scales
A Practical Approach to Sedation Scales and Discharge Scores
Conclusion
References
6: Physiological Monitoring for Procedural Sedation
Introduction
Current Guidelines and Standards
Observational Monitoring
Electronic Monitoring
Oxygenation Monitoring
Ventilation Monitoring
Hemodynamic Monitoring
Cortical Activity Monitoring
Bispectral Index Monitoring
Cerebral Oximetry
Future Directions
Bioacoustic Methods
Noninvasive Cardiovascular Monitoring
Conclusion
References
7: Neuromonitoring and Sedation; Is There a Role?
Introduction: The History
Awareness Under Anesthesia and Neuromonitors
A Global Review of Published Guidelines on Depth of Anesthesia Monitoring
The Range of Neuromonitors: The Science, Practice, and Function
BIS
Validity of BIS Scores with Sedation Scales and Depth of Sedation in Infants and Children
Validation of BIS with Inhalational Agents and Sedatives
Nitrous Oxide
Chloral Hydrate (CHO)
Ketamine
Propofol
Dexmedetomidine
Midazolam
Narcotrend
M-Entropy
Auditory Evoked Potential (AEP)
Limitations of EEG-Based Monitors
Recent Advances and Future Considerations
Conclusion
Case 1
Considerations
Case 2
Considerations
Case 3
Considerations
References
8: The Pediatric Airway: Anatomy, Challenges, and Solutions
Anatomy of the Pediatric Airway
Assessment of the Pediatric Airway for Sedation
Risk Factors for Airway Compromise or Depression
Sleep-Disordered Breathing
Identification and Treatment of Airway-Related Adverse Events
Conclusion
Case Studies
Case 1: Obstructive Sleep Apnea
Case 2: Anterior Mediastinal Mass
Case 3: The Child with a “Cold”
References
9: Pediatric Physiology: How Does It Differ from Adults?
Introduction
Respiratory Physiology
Cardiovascular Physiology
Development from Neonate to Older Infant and Child
Innervation of the Heart
Development from Child to Adult
Normal Heart Rate and Blood Pressure Ranges at Different Ages
CNS Physiology
Hematologic System Development
Renal Physiology and Fluid and Electrolytes
Hepatic/Gastrointestinal Physiology
Temperature Regulation
Drug Pharmacokinetics and Pharmacodynamics
Conclusion
References
10: Capnography: The Science, Logistics, Applications, and Limitations for Procedural Sedation
Why Should Capnography Be Used in Pediatric Patients
Measurement of Exhaled Carbon Dioxide
Side Stream Capnography
Mainstream Capnography
Colorimetric Devices
Physiology of Capnography
Time Capnogram
Volume Capnogram
Arterial to End-Tidal PCO2 Difference
Abnormal Capnograms and Clinical Considerations
Capnograms During Sedation
Sampling Devices
Algorithm for Monitoring Ventilation Via Capnography
Capnography in Dental Practice
Capnography and Cardiopulmonary Resuscitation (CPR)
Conclusion
Case Studies
Case 1
Considerations
Case 2
Considerations
Case 3
Considerations
References
11: Clinical Pharmacology of Sedatives, Reversal Agents, and Adjuncts
Drug Selection and Administration
Use of Multiple Drugs for Sedation
Additional Pharmacologic Effects
Off-Label Use
Alternate Sites of Administration
Reversal Agents
The Effects of Psychotropic Drugs on the Developing Brain
Formulary
Sedatives and Analgesics
Alfentanil (Alfenta, Rapifen)
Chloral Hydrate
Codeine
Dexmedetomidine (Precedex, Dexdor)
Diazepam (Valium, Antenex)
Etomidate (Amidate)
Etomidate Analogs
Fentanyl (Fentanil, Sublimaze, Actiq, Durogesic, Duragesic, Fentora, Onsolis, Instanyl, Abstral)
Fospropofol (Lusedra)
Ketamine (Ketanest, Ketaset, Ketalar)
Ketofol (Ketamine + Propofol)
Lorazepam (Ativan, Temesta)
Meperidine (Demerol, Isonipecaine, Lidol, Pethanol, Piridosal, Algil, Alodan, Centralgin, Dispadol, Dolantin, Mialgin, Petidin Dolargan, Dolestine, Dolosal, Dolsin, Mefedina)
Methohexital (Methohexitone, Brevital)
Midazolam (Versed, Dormicum, Hypnovel)
Morphine (MS Contin, MSIR, Avinza, Kadian, Oramorph, Roxanol, Kapanol)
Nitrous Oxide
Pentobarbital (Nembutal)
Propofol (Diprivan)
Remifentanil (Ultiva)
Remimazolam (Byfavo)
S-Ketamine (Ketanest, Ketaset, Ketalar)
Sufentanil (Sufenta)
Reversal Agents
Flumazenil (Flumazepil, Anexate, Lanexat, Mazicon, Romazicon, Anexate)
Naloxone (Narcan, Nalone, Narcanti)
Local Anesthetics
Lidocaine (Lignocaine)
Anti-emetics
Ondansetron (Zofran)
Metoclopramide (Maxolon, Reglan, Degan, Maxeran, Primperan, Pylomid, Cerucal, Pramin)
Scopolamine (Levo-Duboisine, Hyoscine)
Diphenhydramine (Benadryl, DPH, DHM, Dimedrol, Daedalon)
Dexamethasone
References
Alfentanil
Pharmacokinetics
Chloral hydrate
Pharmacokinetics
Clinical Application
Codeine
Dexmedetomidine
Buccal
Intranasal
Intramuscular
Pharmacokinetics and Pharmacodynamics
Diazepam
Etomidate
Clinical Application
Fentanyl
Transdermal
Intranasal
Transmucosal
Pharmacokinetics
Fospropofol
Ketamine
Pharmacokinetics
Clinical Application
Ketofol
Lorazepam
Pharmacokinetics
Meperidine
Pharmacokinetics
Methohexital
Midazolam
Pharmacokinetics
Morphine
Pharmacokinetics
Nitrous Oxide
Pharmacokinetics
Clinical Application
Pentobarbital
Pharmacokinetics
Propofol
Pharmacokinetics
Clinical Application
Remifentanil
Pharmacokinetics
Clinical Application
Remimazolam
S-Ketamine
Intranasal
Caudal Block
Rectal
Pharmacodynamics and Pharmacokinetics
Sufentanil
Pharmacokinetics
Flumazenil
Intranasal
Rectal
Pharmacokinetics
Naloxone
Pharmacokinetics
Lidocaine
Pharmacokinetics
Ondansetron
Pharmacokinetics
Metoclopramide
Pharmacokinetics
Scopolamine
Diphenhydramine
Pharmacokinetics
Dexamethasone
Pharmacokinetics
12: The Pharmacology, Physiology and Clinical Application in Dentistry of Nitrous Oxide
Introduction
Administration Technique in Dentistry
Monitoring
Contraindications
Adverse Effects
Mechanisms of Action for Nitrous Oxide
Personnel Safety
References
13: Sedation of the Obese Child: Essential Considerations
Introduction and Background
Definition of Obesity
Obesity and Comorbidities
The Pathophysiology of Obesity
Assessment of Obese Children Before Procedural Sedation
Adverse Respiratory Events
Airway Management
Pharmacological Considerations on Obese Children
Pharmacokinetics
Absorption
Distribution
Elimination
Hepatic Metabolism
Renal Elimination
Pharmacodynamics
Drugs and Doses for Sedation of the Obese Child
Sedative/Hypnotics
Thiopental
Propofol
Opioids
Morphine
Fentanyl and Fentanyl Derivatives (Alfentanil, Sufentanil, and Remifentanil)
Dexmedetomidine
Benzodiazepines
Midazolam
Newer Sedatives/Hypnotic Drugs
Conclusion
Case 1
Considerations
Case 2
Considerations
Case 3
Considerations
References
14: Sedation; Is it Sleep, Is it Amnesia, What’s the Difference?
Introduction: Sedation, Sleep, Memory, and Amnesia
Sedation vs. Anesthesia
To Sleep, Perchance To….To Sedate
To Sedate … Perchance … Not to Remember!
Memory: What Is It, Really?
The Last Building Block of a Conscious Memory: Consolidation
The First Building Block of Amnesia: Forgetting
Mechanistic Implications of Drug-Induced Amnesia
Memories We Don’t Know We Have: The Unconscious Mind
Clinical Practice
Sedative Agents: Brief Considerations
Propofol
Benzodiazepines
Dexmedetomidine
Ketamine
Etomidate
Inhalational and Miscellaneous Agents
Case Studies
Case 1
Case 2
Case 3
References
15: Pharmacokinetics and Pharmacodynamics in the Pediatric Population
Introduction
PK Differences in the First Year of Life
Input
Absorption
Enteral
Intramuscular
Nasal
Cutaneous
Alveolar
Bioavailability
Distribution
Body Composition
Plasma Proteins
Regional Blood Flows
Blood–Brain Barrier
Drug Metabolism
Descriptors for Metabolism Maturation
Hepatic Elimination
Phase 1
Phase 2
Renal Elimination
Pulmonary Elimination
Metabolites
Pharmacogenomics
Pharmacodynamic Differences in the First Year of Life
Measurement of PD Endpoints
Population Modeling
The Target Concentration Approach
Pharmacokinetic Models
Pharmacodynamic Models
The Sigmoid Emax Model
Quantal Effect Model
Logistic Regression Model
Linking PK with PD
Adverse Effects
Drug Interactions
Defining Target Concentration
Conclusion
References
16: Billing and Reimbursement for Sedation Services in the United States
Introduction
Minimal Sedation
Moderate Sedation
Key Components of Evaluation and Management Services Documentation
Deep Sedation (MAC)
General, Regional, and Monitored Anesthesia Care
Fee for Services
Physician Quality Measure Reporting
Legal Consequences of Incorrect Coding/Documentation
Conclusion
Case Scenarios
References
Part II: Sedation Models Delivered by Different Specialties: A Global Voyage
17: The Pediatric Hospital Medicine Service: Models, Protocols, and Challenges
Introduction
Training Hospitalists to Provide Moderate and Deep Sedation
On-the-Job Training
Residency and Fellowship Training
Training Under Direct Supervision
Operating Room Time
Simulation Time
Pediatric Advanced Life Support Training
Ongoing Competency
Credentialing Hospitalists to Provide Moderate and Deep Sedation in the United States
Logistics of Setting Up a Hospitalist-Run Sedation Service
Staffing
Staffing Example
Triaging Patients to Sedation by Pediatric Hospitalists
How and When Medical Evaluations Are Performed for Triage
Funding Pediatric Hospitalist Sedation Programs
The Future of Hospitalist Sedation Services
Developing National Standards for Training and Credentialing Pediatric Hospitalists in Sedation
Planning, Monitoring, and Recovering from a Sedation
Pre-sedation Evaluation
Personnel
Monitoring
Medications
Final Checklist Prior to Sedation
Recovery
Discharge/Transfer Criteria
Commonly Administered Sedation Drugs
Ketamine
Dosing
Fentanyl and Midazolam
Dosing
Nitrous Oxide
Dosing
Pentobarbital
Dosing
Propofol
Dosing
Chloral Hydrate
Dosing
Dexmedetomidine
Dosing
Case Scenarios
Case 1
Considerations for Case 1
Sedative Options and Considerations for Case 1
Case 2
Considerations for Case 2
Sedative Options and Considerations for Case 2
For This Case
Case 3
Considerations for Case 3
Sedative Options and Considerations for Case 3
Case 4
Considerations for Case 4
Sedative Options and Considerations for Case 4
Case 5
Considerations for Case 5
Sedation Options and Considerations for Case 5
References
18: Sedation in the Neonatal Intensive Care Unit: International Practice
Introduction
Why Do Neonates Need Procedural Analgo-sedation?
Take-Home Messages: Why a Focused Chapter on Neonatal Analgo-sedation?
Assessment of Distress and Pain in Neonates
Limitations of Assessment of Distress and Pain in Neonates
Implementation of Assessment
Take-Home Messages on Pain Assessment
Preventive Strategies
Take-Home Messages
Complementary Interventions
Non-nutritive Sucking, Sucrose, Glucose, and Human Milk
Swaddling and Containment Procedures
Multisensorial Stimulation and Sensorial Saturation
From Evidence to Practice: The Implementation Issue
Take-Home Messages
Pharmacological Interventions
Topical and Local Anesthesia
Take-Home Messages
Propofol
Take-Home Messages
Ketamine
Pharmacokinetics
Pharmacodynamics
Take-Home Messages
Remifentanil
Take-Home Messages
Chloral Hydrate
Take-Home Messages
Morphine and Fentanyl
Take-Home Messages
Benzodiazepines
Take-Home Messages
Dexmedetomidine
Take-Home Messages
Inhalational Agents
Take-Home Messages
Acetaminophen (Paracetamol)
Efficacy
Safety
Take-Home Messages
Neonatal Analgo-sedation: Balancing Between Scylla and Charybdis
Case studies
Case 1
Issues
Case 2
Potential Options, To Consider
Case 3
Case 4
References
19: Sedation in the Pediatric Intensive Care Unit: Challenges, Outcomes, and Future Strategies in the United States
Introduction
Pre-procedure Preparation
Assessing the Depth of Sedation
Basic Principles
Choice of Agent and Route of Delivery
Inhalational Anesthetic Agents
Benzodiazepines
Etomidate
Ketamine
Propofol
Barbiturates
Opioids
Phenothiazines and Butyrophenones
Alpha2-Adrenergic Agonists
Chloral Hydrate
Tolerance, Physical Dependency, and Withdrawal
Clinical Signs and Symptoms of Withdrawal
Treatment of Withdrawal and Clinical Scoring Systems
Delirium
Classification of Delirium
Diagnosis of Delirium
Pathophysiology of Delirium
Prevention and Treatment of Delirium
Palliative Sedation Therapy
Ketamine
Propofol
Dexmedetomidine
Conclusion
Case Scenarios
Case 1
Considerations
Drugs
Case 2
Considerations
Drugs
Case 3
Considerations
Drugs
References
20: Sedation in the Pediatric Intensive Care Unit: Current Practice in Europe
Introduction
Pharmacologic Aspects
General Aspects
Specific Aspects
Opioids
Benzodiazepines
Ketamine
Inhalational Agents
Alpha-2 Receptor Agonists
Propofol
Immunity
Neuropathologic Effects
Measuring Sedation
Delirium
Tolerance and Withdrawal
Staff
Conclusion
Clinical Case Samples
Case 1
Consideration
Withdrawal
Case 2
Considerations
Delirium
Sleep Cycle
Neuromyopathy
Case 3
Consideration
Context-Sensitive Half-Life
References
21: Sedation for Pediatric Gastrointestinal Procedures
Introduction
Goals and Optimal Levels of Sedation to Minimize Complications for Pediatric GI Procedures
Pre-procedure Preparation and Patient Assessment
Patient Positioning
Common IV Sedation Regimens for Pediatric Gastrointestinal Procedures
Fentanyl
Midazolam
Reversal Agents for Narcotics and Benzodiazepines
Ketamine
Nitrous Oxide
Diprivan (Propofol)
Non-anesthesiologist-Administered Propofol Sedation (NAAPS)
Dexmedetomidine
Training in Sedation Administration
Monitoring of Children Undergoing Endoscopic Procedures with Sedation
Pulse Oximetry
Capnography
Electroencephalography Monitoring
Future Sedation Strategies for Endoscopy in Children
Conclusion
Case Studies
Case 1
Considerations
Case 2
Considerations
Case 3
Considerations
References
22: Sedation in the Emergency Department: A Complex and Multifactorial Challenge
Introduction
Why Procedural Sedation and Analgesia (PSA)?
Long-Term Negative Impact of Painful Procedures
When May PSA Not Be Needed?
Nearly Painless Local Anesthesia
Topical Anesthetics
Buffering Injected Lidocaine
Psychological Interventions Reduce Distress and Need for PSA
What Makes PSA in the Emergency Department Different?
Deciding Whether to Perform PSA
Systematic Approach to Safe ED PSA
Knowledge of Clinical Policies Specific to Emergency Department
Goals of PSA
Pre-sedation Patient Evaluation and Risk Assessment
Informed Consent
Plan for Sedation
Equipment
Preparation for and Management of Adverse Events
Anticipation
Management of Respiratory Depression and Apnea
Airway and Ventilation Maintenance
Treatment: Respiratory Depression and Apnea
First Line (in Rapid Succession, If Needed)
Second Line: Reversal Medications for Opioids and Benzodiazepines
Naloxone (Narcan®)
Flumazenil (Romazicon®)
Upper Airway Obstruction
Treatment
Laryngospasm
Emesis
Treatment: Emesis During Procedural Sedation
Ondansetron (Zofran®)
Pulmonary Aspiration
Medications
Basic Pharmacokinetics: Simplified
Dosing Details
Titration to Desired Effect
Intravenous Administration at the Hub
Intramuscular Administration
Sedative–Hypnotic Agents
Chloral Hydrate [76]
Pregnancy Category C
Barbiturates
Methohexital (Brevital®)
Pregnancy Category B
Pentobarbital (Nembutal®)
Pregnancy Category D
Anxiolytic–Amnestic–Sedative Agents
Benzodiazepines
Paradoxical Reactions
Midazolam (Versed®)
Pregnancy Category D
Diazepam (Valium®)
Pregnancy Category D
Other Non-analgesic Sedative Agents
Propofol (Diprivan®)
Pharmacology
Pharmacokinetics
Pregnancy Category B
Etomidate
Pregnancy Category D
Sedative–Analgesic Agents
Opiates (Narcotics) (Table 22.7): Fentanyl (Sublimaze®)
Pregnancy Category C
Morphine
Pregnancy Category C
Meperidine (Demerol®)
Pregnancy Category C
Codeine
Oxycodone
Pregnancy Category B (D for Prolonged Use)
Hydrocodone
NMDA Antagonists
Ketamine (Ketalar®)
Circulatory Effects
Ventilatory Effects
Protective Airway Reflexes
Sedative–Analgesic Effects
Dissociative Effects
Prolonged Analgesic Effects
Neurotoxicity
Psychotomimetic Effects
Beneficial Psychiatric Effects
Other Adverse Effects
Pharmacokinetics
Pregnancy Category B
Adjuncts
Glycopyrrolate (Robinul®)
Atropine
Ketamine with Sedatives or Analgesics
Ketamine Plus Propofol (Ketofol)
Ketamine Plus Dexmedetomidine (Ketadex)
Nitrous Oxide (N2O)
Pregnancy Category C
Dexmedetomidine
Techniques
Pregnancy Category C
Ketamine + Midazolam or Fentanyl + Midazolam Techniques for Deep Sedation
Pre-sedation Assessment and Preparation
During Sedation
Fentanyl Technique
Ketamine Technique
Conclusion: Final Thoughts
Case Studies
Case 1
Case 2
Case 3
Case 4
Case 5
Case 6
Case 7
References
23: Sedation for Radiological Procedures
Introduction
Radiology Sedation: Common Themes
Magnetic Resonance Imaging (MRI)
Medications
Dexmedetomidine
Propofol
Barbiturates
Chloral Hydrate
Special Populations: Autistic Patients
Nuclear Medicine (NM) Procedures
Magnetoencephalography (MEG)
Interventional Radiology (IR)
Computed Tomography (CT)
Fluoroscopic Voiding Cystourethrogram (VCUG) and Nuclear Medicine Cystogram
Transthoracic Echocardiogram (TTE)
Conclusion
Case Scenarios
Case 1
Considerations
Case 2
Considerations
Case 3
Considerations
References
24: Sedation of Pediatric Patients for Dental Procedures: The USA, European, and South American Experience
Introduction
Section 1
Extent and Treatment of Dental Caries
Children’s Behavior
Measuring Anxiety in Children Undergoing Dental Treatment
Pediatric Dental Sedation Appointment Protocols
Sedatives Used in Pediatric Dentistry
Propofol for Adolescents
The Evidence for Conscious Sedation in Pediatric Dentistry: Cochrane
Morbidity and Mortality: Dental Sedation
Alternatives to Sedation
Section 2
Guidelines and Training in the USA
Sedation Guidelines in the USA
Reimbursement for Dental Sedation and Anesthesia
Future of Sedation for Dental Procedures in the USA
Section 3
Sedation for Pediatric Dental Patients in the UK and Europe
UK Pediatric Dentistry
Background
UK: Local Anesthesia
The UK Definition of Conscious Sedation
National Institute for Health and Care Excellence
NICE Levels of Sedation Definitions
UK Pediatric Dental Sedation Training
Pediatric Dental Sedation and General Anesthesia in the UK
The Link Between “Conscious Sedation” and General Anesthesia
Premedication (Sedation) Prior to General Anesthesia
Titrated Nitrous Oxide Inhalation Sedation
Intravenous Sedation
Oral Sedation
Sedation and Dental-Specific Guidelines of the UK and European Union
British Society of Paediatric Dentistry
European Association of Pediatric Dentistry
Section 4
Pediatric Dental Sedation in South America
What Is the Regulatory Language Related to Performing Dental Sedation in the Pediatric Dental Office?
What Sedation Guidelines Do Pediatric Dentists Follow in South America?
What Sedation Regimens Have Been Used and/or Investigated in South America?
Closing Thoughts on Sedation in South America
Conclusion
The Future
Section 5
Case Studies
Case 1 (from Brazil)
Case 2 (from Brazil)
Case 3
Case 4
Case 5
Case 6
Case 7
Case 8
References
25: Sedation Strategies and Techniques for Painful Dental Procedures
Intraoral Local Anesthetic Techniques
Maxillary Local Anesthesia
Mandibular Local Anesthesia
Specific Dental Equipment and Safety Considerations
Procedural Sedation Techniques and Considerations
Monitoring with Special Considerations to Dental Procedural Sedation
Type of Procedures and Considerations
Dental Extractions
Cleft Lip and Palate Surgeries
Oral Trauma
Postoperative Analgesia
Conclusion
References
26: Special Considerations During Sedation of the Child with Autism Spectrum Disorder
Introduction
Background and Diagnosis
Medical and Psychiatric Comorbidities
Behavioral Challenges
Communication Challenges
Pre-procedural Planning
Patient Selection
Premedication
Intraprocedure Considerations
Conclusion
Case 1
Considerations
The MTHFR Mutation Debate
Our Patient Has a MTHFR Mutation
Our Patient Was Never Tested for a MTHFR Mutation
Case 2
Considerations
Sedation
Debrief
References
27: Pediatric Sedation: The European Experience and Approach
Introduction
General Problems and Challenges
Growing Demand for Optimal Procedural Sedation and Analgesia
Expanding the Horizon: PSA as a Component of Family-Centered Procedural Comfort Care
Defining High-Quality Procedural Sedation
Cultural Aspects, Diversities, and Inconsistencies Within Europe
Anesthesia Services Are Limited in Europe
Non-anesthesiology Sedation Providers in Europe
Challenges and Solutions in Europe
Monitoring Practices in Europe Are Inconsistent
Capnography
Processed EEG
Recommendations, Policy Statements, and Guidelines in Europe
Ethical and Legal Aspects and the Debate on Neurotoxicity
Definitions Particular to Europe
Training and Credentialing in Europe
Implementation of Practice Standards in Europe
Financial Aspects of Sedation Delivery in Europe
Common European Sedation Practice for Selected Procedures
Painless Imaging
Interventional Radiology and Cardiology
Gastroenterology
Oncology
Emergency Medical Care
Dentistry
Conclusion
New and Future Developments
Case Studies
Case 1 (the Netherlands)
The Sedation
Summary Points
Case 2 (the Netherlands)
The Sedation
Summary Points
Case 3 (the Netherlands)
The Sedation
Summary Points
Case 4 (the United Kingdom)
A 6-Year Old Boy for a Surveillance MRI Scan of His Brain
Case 5 (the United Kingdom) [81]
Magnetic Response Imaging (MRI) in a 10-Month-Old
The Sedation
Case 6 (the United Kingdom)
A 13-Year-Old Boy with Down Syndrome and Leukemia
The Sedation
Case 7 (the United Kingdom)
Gastrointestinal Endoscopy in an Anxious 15-Year-Old Girl
References
28: Pediatric Sedation in South America
Introduction
Pediatric Sedation in South America: A General Overview
Argentina
Brazil
Chile
Other Countries
Common Sedation Techniques and Strategies in South America
Review of Published Sedation Literature from South America
Summary
Case Studies
Case 1
Case 2
Case 3
Case 4
References
29: Paediatric Sedation: The Asian Approach—Current State of Sedation in China
Introduction
An Example of Sedation Services in Large-Scale Sedation Unit
Chloral Hydrate
Intranasal Dexmedetomidine
Sedation Service at the Hong Kong Children’s Hospital
Anaesthesiologist-Administered Sedation Services in Hong Kong
Non-painful Procedures
Case Scenario 1
MRI of Brain and Whole Spine
Sedation for Painful Interventional Radiological Procedures
Pre-sedation Assessment
Room Preparation
Choices of Drugs
Dexmedetomidine
Propofol
Ketamine
Fentanyl
Local Anaesthetics (LA)
Other Analgesic Adjuncts
Case Scenario 2
Renal Biopsy
Case Scenario 3
Pleural Mass Biopsy
Electronic Clinical Information System in Sedation Service
References
30: Pediatric Sedation: The Approach in Australia and New Zealand
Introduction
The Key Guiding Documents
The Australian and New Zealand College of Anaesthetists Guideline
Limitation of the ANZCA Guidelines
The Royal Australasian College of Physicians Guideline Statements
Development of a Sedation Program
Specific Locations and Services
Inpatient Wards
Sedation in the Pediatric Intensive Care Units
Neonatal Intensive Care Units
Emergency Departments
Dental Sedation
Sedation for Medical Imaging
Sedation of Children with Burns
Sedation for Oncologic Procedures
Non-pharmacological Management
Case Studies
Case 1
Case 2
Case 3
References
31: Pediatric Sedation in the Underdeveloped Third World: An African Perspective
Introduction
Provision of Safe Pediatric Sedation in the Third World
Recovery and Discharge
Discharge from the Facility to Home
Sedation Models
The Sedation Unit Model Within the Hospital
The Mobile Sedation Model Within the Hospital
A Combined Sedation Model Within the Hospital
The Mobile Sedationist Model Outside the Hospital
The Operator-Sedationist Model
The Dedicated Mobile Sedationist Model
Common Sedation Strategies in the Developing Nations
Oral Route: Single Agent
Nasal Route: Single Agent
Rectal Route: Single Agent
Intravenous Route: Single Agent
Inhalational Route
Sedative and Analgesic Combination
Behavioral and Non-pharmacological Method
Sedation Training Opportunities in Africa
Accreditation of Providers and Clinics
Clinical Governance in South Africa
Patient Satisfaction in Children
Conclusion
Case Studies
Case 1
Case 2
Case 3
References
Part III: Safety in Sedation
32: Pediatric Sedatives and the Food and Drug Administration (FDA): Challenges, Limitations, and Drugs in Development
Introduction
General Drug Development
Pediatric Legislation
Drug Development for Pediatrics
Chemistry, Manufacturing, and Controls
Nonclinical Studies
Clinical Trials
Ethics
Pharmacokinetics and Pharmacodynamics
Pediatric Extrapolation
Phase 3 Safety and Efficacy Studies
References
33: Apoptosis and Neurocognitive Effects of Intravenous Anesthetics
Introduction
Characterization of Sedative-Induced Developmental Neurotoxicity
Mechanisms of Aberrant Neuronal Development from Sedative Drugs
Clinical Evidence for Sedative-Induced Neurological Sequelae
Conclusions from Preclinical and Clinical Investigations
References
34: Adverse Events: Risk Factors, Predictors, and Outcomes
Introduction
Patient Characteristics
Age/Prematurity
Weight/Obesity/Underweight
Genetic/Trisomy 21/Metabolic
Autistic Spectrum Disorder
Cerebral Palsy
Congenital Heart Disease/Pulmonary Hypertension
Cancer/Mediastinal Mass
Nil Per Os (NPO) Status
URI Status
Current Fever
Allergy
Procedural Characteristics
Airway Manipulation: Dental, Bronchoscopy, and Esophagogastroduodonescopy (EGD)
Invasive vs Noninvasive
Positioning: Supine vs Prone
Duration
Emergent vs Urgent vs Elective
Environmental Characteristics
Indirect Patient Access: CT, MRI, and Radiation
Remote Locations within Hospital Locations
Satellite Locations
Free-Standing Sedation Locations
Provider Characteristics
Medications
Predictive Tools
ASA Physical Status Classification
Outcome Studies
Case Scenarios
Case 1
Considerations
Case 2
Considerations
Case 3
Considerations
References
35: Fasting Status, Aspiration Risk, and Sedation Outcomes
Introduction
Incidence and Severity of Clinically Apparent Pulmonary Aspiration
Aspiration Risk in Procedural Sedation Compared to General Anesthesia
Fasting Status as a Risk Factor for Aspiration
Disadvantage of Fasting Guidelines
Specialty Society Recommendations for Fasting Prior to Procedural Sedation
Additional Risk Factors for Aspiration
Strategies to Mitigate Aspiration Risk
Gastric Ultrasound
Selection of Sedation Medication
Duration and Depth of Sedation
Referral to Anesthesiology
ICAPS Strategy
Conclusions and Future Directions
Case Studies
Case 1
Considerations
Case 2
Considerations
Case 3
Considerations
References
36: Outcomes of Procedural Sedation: What Are the Benchmarks?
Background
Introduction
Setting the Standards for Safety
Safety: Disparities in Adverse Event Rate Reporting
Safety: Recommendations for Defining and Reporting Adverse Events
Efficacy of Sedation
Tracking and Reporting Outcomes of Procedural Sedation
Multicenter Investigations
Future Directions
Conclusions
References
37: Medicolegal Risks and Outcomes of Sedation
Introduction and Background
Preventing Litigation
Practice “Good” Medicine
The Sedation Process
Pre-sedation Evaluation/Decisions
Medication Errors
Post-sedation/Discharge
Policies and Protocols
Clinical Guidelines
Communicate Well with Patients/Families
Informed Consent
Communicate Well with Colleagues
Document Carefully
Never Alter Medical Records
Managing Medical Errors in the Event of an Adverse Event
When to Contact an Attorney
Quality Improvement
Family Member Presence for Procedures
Case Studies
Case 1
Case 2
Case 3
Case 4
Case 5
Case 6
Conclusion
Glossary
References
Additional Reading
38: Improving the Safety of Pediatric Sedation: Human Error, Technology, and Clinical Microsystems
Introduction
The Need for a Paradigm Shift
Errors and Violations
The Nature of Medication Error in Children
Additional Causes of Errors in Children
The Clinical Microsystem as a Unit of Analysis
People Versus Systems
Rules and Rule Following: The Boeing MAX Aircraft and the Thailand Cave Rescue
Making Sense of Uncommon Adverse Events
The Nature of Human Error
Error Types
Knowledge-Based Errors (or Errors of Deliberation)
Rule-Based Errors
Skill-Based Errors
Technical Errors
Exhortation and Protocols
The Effects of Fatigue
Human Factors and the Culture of Safety
Simulation and Safety
Teamwork and Communication
The Nature of System Failures
Characteristics of Safe and Unsafe Systems
Barriers to System Failure
Traversing the Incident Pyramid
Root Cause Analysis
Failure Mode and Effect Analysis
Lessons from the Development of Safety in the Nuclear Power Industry
The Three Mile Island Nuclear Power Plant Accident
A State-Space Approach to Failure in Complex Systems
The Role of Incident Reporting
The Value of Incident-Based Recovery Pathways
Closed Claims Settlements for Cases Outside the Operating Room
The Value of Best Practice Guidelines in Procedural Sedation
The Future of Safety in Pediatric Sedation
New Approaches to Safety Monitoring and Improvement
Conclusion
References
Part IV: Sedation into the Twenty-Second Century
39: Intravenous Infusions for Sedation: Rationale, State of the Art, and Future Trends
Introduction
Benefits of the Intravenous Route of Administration
Choice of Agents
Pharmacodynamics of Commonly Used Agents
Propofol
Dexmedetomidine
Ketamine
Remifentanil
Basic Principles of Pharmacokinetics
What Is a Pharmacokinetic Model and How Is It Derived?
Important Mathematical Concepts for Understanding of Pharmacokinetic Models
Half-Life, Time Constant, and Rate Constant
Volume of Distribution
Single Compartment Pharmacokinetic Models
Three Compartment Models
Context-Sensitive Half-Time
Pharmacokinetic Models for Propofol
Pediatric Propofol Infusion Regimens
Disadvantages of Repeated Bolus Dose Administration
Commonly Used Regimens
PK Models for Dexmedetomidine
Infusion Regimens for Dexmedetomidine
PK Models for Ketamine
Infusion Regimens for Ketamine
PK Models for Remifentanil
Infusion Regimens for Remifentanil
Target-Controlled Infusions
Definition
Rationale for TCI
Principles of TCI
Choice of Propofol Target Concentration
Predictive Performance of PK Models During TCI
Choice of Dexmedetomidine and Remifentanil Target Concentrations
Future Directions
Model Development and the Open TCI Initiative
Drug Interactions
Effect-Site Targeted TCI Systems
Closed-Loop Control
References
40: Usage of Nonpharmacological Complementary and Integrative Medicine in Pediatric Sedation
Introduction
Guided Imagery
Acupuncture and Related Techniques
Music
Hypnotherapy
Sucrose Sucking in Infants
Conclusion
Case Study
Case 1
Suggestions
Case Study
Case 2
Suggestions
Case Study
Case 3
Suggestions
References
41: Towards Integrated Procedural Comfort Care: Redefining and Expanding “Non-pharmacology”
Introduction
The “Non-sense” and “Complexity” of “Non-pharmacology”
Non-pharmacology Versus Pharmacology
“Non-pharmacology”: A Problematic Concept
Non-pharmacology as a Specific Educational Challenge
Deconstructing Non-pharmacology
First of All: Do Not Harm
Creating the Human Connection
Relevance of Previous Experience and Information
Procedural Disclosure May Do Harm Concept
Right Words, Wrong Words
Distraction Versus Reassurance
Role of Parents
Evaluation of the Child Level of Anxiety and Search for the Less Scary Approach
Conclusion
Physical Comfort Measures and Physical Analgesia
Distraction Techniques
Virtual Reality as a Possible New Frontier
Burn Care
Venipuncture
Dental Treatment
Limitations of the Technology
Conclusion
Final Conclusions
References
42: The Role of Simulation in Safety and Training
History of Medical Simulation
General Applications of Simulation for Sedation Safety and Training
Simulation for Pediatric Sedation
Examples of Simulation-Based Training in Pediatric Sedation
Initial Training
Advanced Training
General Principles for the Development of a Pediatric Sedation Simulation Scenario and Course
Pediatric Sedation Simulation Scenario Development
Sample Pediatric Sedation Simulation Scenario
Debriefing
Evaluation of Pediatric Simulation Activities
Future of Simulation for Pediatric Sedation Training
References
43: Criminal Homicide Versus Medical Malpractice: Lessons from the Michael Jackson Case and Others
Legal Versus Professional Standards and Ethics
The Death of Michael Jackson: A Legal Perspective
The Death of Michael Jackson: Professionalism and Medical Ethics
Was Dr. Murray Practicing Medicine?
Did a Legitimate Physician-Patient Relationship Exist?
Ethical Principles in Medical Care
Respect for Autonomy
Beneficence
Nonmaleficence
Outcome of the Case
Another Sedation Case
The Case
Legal Analysis
Professionalism and Ethics
Summary
Glossary
References
44: Considerations for the Intersection of Sedation and Marijuana
Introduction
Pharmacology of Cannabis
Medical Cannabis
Indications
Formulations
Acute Cannabinoid Effects and Withdrawal
Cardiovascular Actions
Pulmonary Actions
Gastrointestinal Actions
Cannabis and Procedural Sedation
Summary
Case 1
Considerations
Case 2
Considerations
Case 3
Considerations
References
45: Proportionate Sedation in Pediatric Palliative Care
Introduction
The Principles of Proportionate Sedation
The Practice of Proportionate Sedation
Pain
Dexmedetomidine (DEX)
Ketamine
Propofol
Dyspnea
Opioids
Dexmedetomidine (DEX)
Delirium
Dexmedetomidine (DEX)
Conclusions
Case Studies
Case #1
Considerations
Case #2
Considerations
Case #3
Considerations
References
46: Ethical and Clinical Aspects of Palliative Sedation in the Terminally Ill Child
Introduction
Palliative Sedation: What Exactly Are We Talking About?
Deep Continuous Sedation
Epidemiology of DCS
What Is Suffering?
Existential Suffering and Transcendence
DCS and Euthanasia: Is Permanent Loss of Consciousness a Form of Death?
What Is Euthanasia?
Defining Death
DCS and Hastening Death
The Principle of Double Effect and DCS
DCS and Withdrawal of Hydration and Nutrition
Legal Precedents and Physician Attitudes Regarding DCS
Professional Societies and Opinions Regarding DCS
Guidelines
Initiating DCS: Goals, Monitoring, and Evaluation of Efficacy
Recommendations for Initiating DCS in Pediatric Patients [77]
Conclusion
Case Studies
Case 1
Considerations
Case 2
Considerations
Case 3
Considerations
References
47: Future of Pediatric Sedation
Introduction
The Optimal Level of Sedation
Outcome Assessment and Standardization of Adverse Event Identification and Documentation
Defining the Depth of Sedation
“Consciousness” Monitoring as an Indication of Sedation Depth
Assessment of Oxygenation, Respiration, and Identification of Hypoxia
Pulse Oximetry
Capnography
Risk Assessment in Balancing the Urgency for the Procedure with the Associated Risk of Sedation
Analgesia, Prophylaxis, and Avoiding Conditioned Behaviors
Training and Credentialing of Sedation Providers
Educating the Public
Developing the “Safety Culture” of Sedation: Implementing Safety Measures
Collecting Outcome Data to Guide Safety and Practice Parameters: Adoption of Standardized Definitions of Sedation-Related Adverse Events
Sedatives: Exploring New Agents and Alternative Methods and Modes of Delivery
Propofol
Etomidate
Alpha 2 Agonists
Synthetic Opioids
Benzodiazepines
Ketamine
Chemical Restraint
Drug Administration
Conclusion
References
Epilogue
Index

Citation preview

Keira P. Mason Editor

Pediatric Sedation Outside of the Operating Room A Multispecialty International Collaboration Third Edition

123

Pediatric Sedation Outside of the Operating Room

Keira P. Mason Editor

Pediatric Sedation Outside of the Operating Room A Multispecialty International Collaboration Third Edition

Editor Keira P. Mason Department of Anaesthesia Harvard Medical School Boston, MA USA

ISBN 978-3-030-58405-4    ISBN 978-3-030-58406-1 (eBook) https://doi.org/10.1007/978-3-030-58406-1 © Springer Nature Switzerland AG 2021 This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or part of the material is concerned, specifically the rights of translation, reprinting, reuse of illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way, and transmission or information storage and retrieval, electronic adaptation, computer software, or by similar or dissimilar methodology now known or hereafter developed. The use of general descriptive names, registered names, trademarks, service marks, etc. in this publication does not imply, even in the absence of a specific statement, that such names are exempt from the relevant protective laws and regulations and therefore free for general use. The publisher, the authors and the editors are safe to assume that the advice and information in this book are believed to be true and accurate at the date of publication. Neither the publisher nor the authors or the editors give a warranty, expressed or implied, with respect to the material contained herein or for any errors or omissions that may have been made. The publisher remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. This Springer imprint is published by the registered company Springer Nature Switzerland AG The registered company address is: Gewerbestrasse 11, 6330 Cham, Switzerland

This book is primarily dedicated to my mother and father, whose sacrifice, love, and encouragement enabled me to pursue my goals and dreams. Leading by example, they showed me to persevere, remain positive, optimistic, and always strive to achieve my personal best. Thank you, God, for blessing me with the gift of my two sons, Tyler and Colin. I will continue to strive to guide, nurture, be a role model for and provide for them, as my parents did for me.

Preface

I am honored and thrilled to present within the relatively short span of only 5 years, this updated and expanded 3rd edition. All 47 book chapters were received within 6 months of book publication – presenting the reader with a completely up-to-date book! The most recent chapters were updated in December, 2020! Every chapter in this book has been updated and there are 12 brand new chapters. All authors are pioneers and world leaders in their areas of expertise, both in the United States and abroad, not only in the field of sedation but also in the areas of law, ethics, child psychology, child development, pediatrics, neonatology, simulation, drug development, patient safety, emergency medicine, intensive (pediatric and neonatal) care medicine, anesthesia, dental anesthesia, dental medicine, psychiatry, neurology, behavioral medicine, and pharmacology. This book is a testimony to the passion and commitment of all the contributing authors to advance the knowledge and practice of pediatric sedation. Pediatric Sedation Outside of the Operating Room: A Multispecialty International Collaboration is intended to represent and be applicable to sedation providers of any specialty from around the world. Our international contributors represent Australia, Belgium, Brazil, Chile, China, Israel, the Netherlands, New Zealand, South Africa, Switzerland, and the United Kingdom. I am very appreciative of their efforts and I extend a sincere “thank you” to each author. There are 12 new chapters in this book, exploring current, evolving, and anticipated relevant topics in sedation. Even the updated chapters, for example, Improving the Safety of Pediatric Sedation: Human Error, Technology, and Clinical Microsystems, is so up-to-date to include not only issues facing the Boeing MAX but also a detailed description of the June 2018 cave rescue in Thailand of the 12 young boys and their coach that were extracted in a water rescue requiring sedation. This chapter is unique in that Dr. Richard Harris, the anesthesiologist from Australia who was on-site and directly involved in orchestrating, directing, and administering the sedation and performing the cave rescue, is one of the authors of this chapter! Additional exciting new chapters that address pertinent new issues include Considerations for the Intersection of Sedation and Marijuana; Behavioral Techniques for Anxiolysis and Sedation; The Autistic Spectrum: Clinical Aspects and Approaches to Sedation; Sedation for End of Life: A Holistic Approach; Fasting Status, Aspiration Risk, and Sedation Outcomes; Neuromonitoring and Sedation; Is There a Role?; Sedation of the Obese Child: Essential Considerations; Adverse Events: Risk Factors, Predictors and Outcomes; and Sedation Strategies and Techniques for Painful Dental Procedures. This book is a unique and authoritative contribution to the field of pediatric sedation. It is directed to all specialties and specifically acknowledges and reviews the contributions and viewpoints of a broad range of international societies and specialists. Sedation has evolved to include all specialties. Although each chapter is written by an expert and world-renowned specialist(s) in his/her particular area, it is intended to be of value to those who do not necessarily practice in that area. For example, the pediatrician in the United States will learn something in the Pediatric Sedation in South America chapter that can be applied or considered for his or her own practice. Chapters specifically written by clinicians and scientists in Asia, Africa, Europe, South America, Australia and New Zealand, explore sedation practice in these countries and continents.

vii

viii

Preface

The chapters that are clinically oriented conclude with Case Studies, which present challenging clinical scenarios. This is a unique finale as it is the author’s presentation of real-life cases. The intent of these Case Studies is to guide the reader through the challenges, thought processes, and management options for each situation. Certainly, there are many possible solutions to each scenario. Exploring them through the eyes of the experienced author offers a unique and valuable perspective. This book may be read cover to cover or read a chapter at a time, out of succession. There is intentional, albeit minimal, repetition in the book. The repetition is intended not only to solidify important information for the reader, but also to convey relevant information for those who may not be reading the book cover to cover. Even the “repetition” is presented in a different style by the individual authors, in most cases masking the repeated elements. The final form of this book went to the publisher in January, 2021. Every chapter was updated in even those final weeks with any recently published papers. This book represents a global collaboration. Currently, the field of sedation is being challenged by politics, differing viewpoints and our inability to reach a consensus. Our ability to come together, outside of this book, will be essential to the future of our pediatric patients who receive sedation. There will continue to be new clinical and research studies that advance our knowledge of sedation. New sedatives, physiological monitors, and sedation delivery systems will certainly be introduced over the next decade. Regardless, the approach to sedation and the information conveyed in these chapters is intended to distinguish this book as a timeless relic that marks an important era in the field of sedation.

Boston, MA, USA January 11, 2021

Keira P. Mason

Acknowledgments

The most important and heartfelt acknowledgment is to my family. Thank you to my two sons, Colin and Tyler, for accepting some “motherless” weekends and evenings throughout my career, as I traveled and shared my passion for and knowledge of sedation with colleagues worldwide. You have, albeit unknowingly, indirectly supported me not only for this book but also throughout my career: Understanding that sharing sedation experience with others to advance the practice, safety, and knowledge of sedation is my passion and has taken me away from home, even missing on occasion some of your important events. Thank you, Tyler, for sitting beside me in the early morning and late-night hours to read over my shoulder, encouraging and helping me edit and revise chapters. Thank you, Colin, for your efforts in helping me organize the book and come up with new ideas for book chapters. I am so proud of you both and honored to be your mother.

I would like to express my respect, gratitude, and appreciation to Gregory Sutorius, Senior Editor for Clinical Medicine, and Lorraine Coffey, Developmental Editor, at Springer. Thank you both for agreeing to the short deadline from inception of the third edition, to receiving the chapters, editing them, formatting the ix

x

book, and presenting it published in 6 short months! Your gentle prodding, attention to detail, kindness, expertise, and professionalism has inspired me to meet all deadlines. Most importantly, you both were committed to this project committed to supporting all efforts to produce Pediatric Sedation Outside of the Operating Room as a contribution to the field. My final acknowledgment is to Ms. Kimberly Manning. From the inception of the 3rd edition of this book to the final moments of its galley proof approval, you devoted even the after-­hours ensuring that all references, figures, tables, and source information were accurate, the grammar and typos corrected, the copyrights were obtained, and that everything from the table of contents to the final chapter flowed appropriately. You caught mistakes that would have gone unnoticed. I have esteem for your commitment to this edition: your organization, encouragement, tireless enthusiasm, and sleuthing skills to uncover important contributions to this book were invaluable. I will always be appreciative.

Acknowledgments

Contents

Part I Pediatric Sedation Outside the Operating Room 1 The History of Sedation���������������������������������������������������������������������������������������������   3 Robert S. Holzman 2 Sedation Policies, Recommendations, and Guidelines Across the Specialties and Continents�����������������������������������������������������������������������������������  21 Joseph P. Cravero 3 Procedural Sedation: Let’s Review the Basics���������������������������������������������������������  41 Vincent W. Chiang and M. Saif Siddiqui 4 Pre-sedation Assessment���������������������������������������������������������������������������������������������  49 Timothy Horeczko and Mohamed Mahmoud 5 Sedation Scales and Discharge Criteria: How Do They Differ? Which One to Choose? Do They Really Apply to Sedation?�����������������������������������  83 Dean B. Andropoulos 6 Physiological Monitoring for Procedural Sedation�������������������������������������������������  95 Cyril Sahyoun and Baruch S. Krauss 7 Neuromonitoring and Sedation; Is There a Role?��������������������������������������������������� 107 Neena Seth 8 The Pediatric Airway: Anatomy, Challenges, and Solutions ��������������������������������� 125 Lynne R. Ferrari 9 Pediatric Physiology: How Does It Differ from Adults?����������������������������������������� 141 Dean B. Andropoulos 10 Capnography: The Science, Logistics, Applications, and Limitations for Procedural Sedation��������������������������������������������������������������������������������������������� 155 Bhavani Shankar Kodali 11 Clinical Pharmacology of Sedatives, Reversal Agents, and Adjuncts ������������������� 171 Randy P. Prescilla 12 The Pharmacology, Physiology and Clinical Application in Dentistry of Nitrous Oxide������������������������������������������������������������������������������������� 199 Dimitris Emmanouil 13 Sedation of the Obese Child: Essential Considerations������������������������������������������ 211 Tom G. Hansen and Thomas Engelhardt 14 Sedation; Is it Sleep, Is it Amnesia, What’s the Difference?����������������������������������� 223 Robert A. Veselis and Vittoria Arslan-Carlon

xi

xii

15 Pharmacokinetics and Pharmacodynamics in the Pediatric Population��������������� 247 Brian J. Anderson 16 Billing and Reimbursement for Sedation Services in the United States ��������������� 273 Devona J. Slater Part II Sedation Models Delivered by Different Specialties: A Global Voyage 17 The Pediatric Hospital Medicine Service: Models, Protocols, and Challenges ����������������������������������������������������������������������������������������������������������� 285 Douglas W. Carlson and Suzanne S. Mendez 18 Sedation in the Neonatal Intensive Care Unit: International Practice ����������������� 305 Karel Allegaert and John van den Anker 19 Sedation in the Pediatric Intensive Care Unit: Challenges, Outcomes, and Future Strategies in the United States ������������������������������������������� 345 Pradip Kamat and Joseph D. Tobias 20 Sedation in the Pediatric Intensive Care Unit: Current Practice in Europe������������������������������������������������������������������������������������������������������� 373 Stephen D. Playfor and Ian A. Jenkins 21 Sedation for Pediatric Gastrointestinal Procedures������������������������������������������������ 397 Jenifer R. Lightdale 22 Sedation in the Emergency Department: A Complex and Multifactorial Challenge������������������������������������������������������������������������������������� 413 Robert M. Kennedy 23 Sedation for Radiological Procedures����������������������������������������������������������������������� 475 Amber P. Rogers 24 Sedation of Pediatric Patients for Dental Procedures: The USA, European, and South American Experience������������������������������������������� 497 Stephen Wilson, Luciane Rezende Costa, and Marie Therese Hosey 25 Sedation Strategies and Techniques for Painful Dental Procedures ��������������������� 533 James W. Tom 26 Special Considerations During Sedation of the Child with Autism Spectrum Disorder ������������������������������������������������������������������������������� 545 John W. Berkenbosch, Thuc-Quyen Nguyen, Dimitris Emmanouil, and Antonio Y. Hardan 27 Pediatric Sedation: The European Experience and Approach������������������������������� 561 Piet L. J. M. Leroy and Grant M. Stuart 28 Pediatric Sedation in South America ����������������������������������������������������������������������� 587 Pablo Osvaldo Sepúlveda and Paulo Sérgio Sucasas da Costa 29 Paediatric Sedation: The Asian Approach—Current State of Sedation in China��������������������������������������������������������������������������������������������������� 601 Vivian Man Ying Yuen, Bi-Lian Li, Bin Xue, Ying Xu, Jacqueline Cheuk Kwun Tse, and Rowena Sau Man Lee 30 Pediatric Sedation: The Approach in Australia and New Zealand������������������������� 615 Franz E. Babl, Ian McKenzie, and Stuart R. Dalziel 31 Pediatric Sedation in the Underdeveloped Third World: An African Perspective����������������������������������������������������������������������������������������������� 633 James A. Roelofse, Graeme S. Wilson, and Cherese Lapere

Contents

Contents

xiii

Part III Safety in Sedation 32 Pediatric Sedatives and the Food and Drug Administration (FDA): Challenges, Limitations, and Drugs in Development���������������������������������������������� 647 Lisa L. Bollinger and Lynne P. Yao 33 Apoptosis and Neurocognitive Effects of Intravenous Anesthetics ����������������������� 657 Sulpicio G. Soriano and Laszlo Vutskits 34 Adverse Events: Risk Factors, Predictors, and Outcomes������������������������������������� 665 Kevin G. Couloures and James H. Hertzog 35 Fasting Status, Aspiration Risk, and Sedation Outcomes��������������������������������������� 681 Maala Bhatt 36 Outcomes of Procedural Sedation: What Are the Benchmarks?��������������������������� 695 Mark G. Roback 37 Medicolegal Risks and Outcomes of Sedation��������������������������������������������������������� 707 Steven M. Selbst and Stewart L. Cohen 38 Improving the Safety of Pediatric Sedation: Human Error, Technology, and Clinical Microsystems ������������������������������������������������������������������� 721 Craig S. Webster, Brian J. Anderson, Michael J. Stabile, Simon Mitchell, Richard Harris, and Alan F. Merry Part IV Sedation into the Twenty-Second Century 39 Intravenous Infusions for Sedation: Rationale, State of the Art, and Future Trends������������������������������������������������������������������������������������������������������� 755 Anthony R. Absalom 40 Usage of Nonpharmacological Complementary and Integrative Medicine in Pediatric Sedation ��������������������������������������������������������������������������������� 773 Yuan-Chi Lin 41 Towards Integrated Procedural Comfort Care: Redefining and Expanding “Non-­pharmacology”���������������������������������������������������������������������� 783 Cyril Sahyoun, Giorgio Cozzi, Piet L. J. M. Leroy, and Egidio Barbi 42 The Role of Simulation in Safety and Training ������������������������������������������������������� 797 James J. Fehr, Itai M. Pessach, and David A. Young 43 Criminal Homicide Versus Medical Malpractice: Lessons from the Michael Jackson Case and Others������������������������������������������������������������� 813 Gail A. Van Norman and Joel S. Rosen 44 Considerations for the Intersection of Sedation and Marijuana��������������������������� 827 Brian E. McGeeney and Rachael Rzasa Lynn 45 Proportionate Sedation in Pediatric Palliative Care����������������������������������������������� 835 Jason Reynolds 46 Ethical and Clinical Aspects of Palliative Sedation in the Terminally Ill Child����������������������������������������������������������������������������������������� 847 Gail A. Van Norman 47 Future of Pediatric Sedation ������������������������������������������������������������������������������������� 863 James R. Miner Epilogue������������������������������������������������������������������������������������������������������������������������������� 881 Index������������������������������������������������������������������������������������������������������������������������������������� 889

Contributors

Anthony R. Absalom, MD, MB, ChB  Department of Anesthesiology, University Medical Center Groningen, Groningen, The Netherlands Karel Allegaert, MD, PhD  Department of Development and Regeneration, and Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands Department of Development and Regeneration, KU Leuven, Neonatal Intensive Care Unit, Leuven, Belgium Brian J. Anderson, PhD  Department of Anaesthesiology, School of Medicine, University of Auckland, Auckland, New Zealand Dean  B.  Andropoulos, MD, MHCM Department of Anesthesiology, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX, USA John van den Anker, MD, PhD  Department of Pediatrics, Pharmacology and Physiology, Children’s National Medical Center, Washington, DC, USA Intensive Care, Erasmus Medical Center-Sophia Children’s Hospital, Rotterdam, The Netherlands Department of Pediatric Pharmacology, University Children’s Hospital Basel, Basel, Switzerland Vittoria  Arslan-Carlon, MD Department of Anesthesiology and Critical Care Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA Franz E. Babl, MD, MPH  Emergency Department, Royal Children’s Hospital, Melbourne, VIC, Australia University of Melbourne, Melbourne, VIC, Australia Murdoch Children’s Research Institute, Melbourne, VIC, Australia Egidio Barbi, MD  Department of Pediatrics, Institute for Maternal and Child Health – IRCCS ‘Burlo Garofolo’, Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy John W. Berkenbosch, MD, FAAP, FCCM  Pediatrics/Pediatric Critical Care, University of Louisville, Norton Children’s Hospital, Louisville, KY, USA Maala  Bhatt, MD, MSc Division of Emergency Medicine, Department of Pediatrics, Children’s Hospital of Eastern Ontario, University of Ottawa, Ottawa, ON, Canada Lisa L. Bollinger, MD  Global Patient Safety and Pediatrics, Global Regulatory Affairs and Safety, Amgen Inc, Thousand Oaks, CA, USA Douglas W. Carlson, MD  Southern Illinois University School of Medicine, Department of Pediatrics, Pediatrics and Emergency Medicine, Springfield, IL, USA xv

xvi

Vincent  W.  Chiang, MD Department of Medicine, Harvard Medical School, Boston Children’s Hospital, Boston, MA, USA Stewart L. Cohen, Esq  Law Offices of Cohen, Placitella & Roth, P.C, Philadelphia, PA, USA Luciane  Rezende  Costa, DDS, MS, PhD Division of Pediatric Dentistry, Universidade Federal de Goias, Goiania, Brazil Kevin G. Couloures, DO, MPH  Division of Pediatric Critical Care Medicine, Department of Pediatrics, Stanford University/Lucille Packard Hospital for Children, Palo Alto, CA, USA Stanford School of Medicine, Palo Alto, CA, USA Giorgio Cozzi, MD  Emergency Department, Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, Italy Joseph P. Cravero, MD  Harvard Medical School, Boston, MA, USA Department of Anesthesiology, Critical Care, and Pain Medicine, Boston Children’s Hospital, Boston, MA, USA Stuart R. Dalziel, MBChB, FRACP, PhD  Departments of Surgery and Paediatrics, Child and Youth Health, University of Auckland, Auckland, New Zealand Children’s Emergency Department, Starship Children’s Hospital, Auckland District Health Board, Auckland, New Zealand Dimitris  Emmanouil, DDS, MS, PhD Department of Pediatric Dentistry, National and Kapodistrian University of Athens, Athens, Greece Thomas  Engelhardt, MD, PhD, FRCA Department of Anesthesia, Montreal Children’s Hospital, Montreal, QC, Canada James J. Fehr, MD  Lucile Packard Children’s Hospital, Palo Alto, CA, USA Stanford University School of Medicine, Stanford, CA, USA Lynne  R.  Ferrari, MD Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston, MA, USA Tom  G.  Hansen, MD, PhD Department of Anesthesiology & Intensive Care: Pediatrics, Odense University Hospital, Odense, Denmark Department of Clinical Research: Anesthesiology, University of Southern Denmark, Odense, Denmark Antonio  Y.  Hardan, MD Division of Child and Adolescent Psychiatry, Autism and Developmental Disorders Clinic, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA Richard Harris, BMBS, D. Univ  Specialist Anaesthetic Services, Adelaide, SA, Australia James  H.  Hertzog, MD Division of Pediatric Critical Care Medicine, Department of Pediatrics, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA Robert S. Holzman, MD, MA (Hon.), FAAP  Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston, MA, USA Timothy  Horeczko, MD, MSCR Department of Emergency Medicine, Harbor—UCLA Medical Center, Torrance, CA, USA Marie Therese Hosey, BDS, MSc  Department of Paediatric Dentistry, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London, UK

Contributors

Contributors

xvii

Ian A. Jenkins, MBBS  Department of Pediatric Intensive Care and Anesthesiology, Bristol Royal Hospital for Children, Bristol, UK Pradip Kamat, MD, MBA, FCCM  Department of Pediatrics, Division of Pediatric Critical Care Medicine, Emory University School of Medicine, Children’s Healthcare of Atlanta, Atlanta, GA, USA Robert  M.  Kennedy, MD Department of Pediatrics, Division of Emergency Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO, USA Bhavani Shankar Kodali, MD  University of Maryland School of Medicine, Baltimore, MD, USA Baruch  S.  Krauss, MD, Ed.M. Division of Emergency Medicine, Boston Children’s Hospital, Boston, MA, USA Cherese  Lapere, MBChB, DipPEC, DA(SA), PDD Department of Anaesthesia and Perioperative Medicine, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa Rowena Sau Man Lee, MBBS, FHKCA  Department of Anaesthesiology and Perioperative Medicine, Hong Kong Children’s Hospital, Hong Kong, China Piet L. J. M. Leroy, MSc, MD, PhD  Pediatric Procedural Sedation Unit, Division of Pediatric Critical Care, Department of Pediatrics, Maastricht University Medical Centre, Maastricht, The Netherlands Bi-Lian Li, MD  Department of Anesthesiology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China Jenifer  R.  Lightdale, MD, MPH Division of Pediatric Gastroenterology and Nutrition, Department of Pediatrics, UMass Memorial Children’s Medical Center, University of Massachusetts Medical School, Worcester, MA, USA Yuan-Chi Lin, MD, MPH  Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA Rachael Rzasa Lynn, MD  Department of Anesthesiology, University of Colorado School of Medicine, University of Colorado Hospital Pain Management Clinic, Aurora, CO, USA Mohamed  Mahmoud, MD Department of Anesthesia, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA Brian  E.  McGeeney, MD, MPH, MBA Division of Headache Medicine, Department of Neurology, Brigham and Women’s Hospital, Brigham and Women’s Faulkner Hospital, JR Graham Headache Clinic, Boston, MA, USA Ian  McKenzie, MBBS, FANZCA  Department of Anaesthesia and Pain Management, The Royal Children’s Hospital, Melbourne, VIC, Australia Suzanne S. Mendez, MD  Department of Pediatrics and Neonatology, St. Charles Medical Center, Bend, OR, USA Alan  F.  Merry, FANZCA, FPMAN  Department of Anaesthesiology, School of Medicine, University of Auckland, Auckland, New Zealand Auckland City Hospital, Auckland, New Zealand James  R.  Miner, MD Department of Emergency Medicine, Hennepin County Medical Center, Minneapolis, MN, USA

xviii

Simon  Mitchell, MBChB, PhD Department of Anaesthesiology, School of Medicine, University of Auckland, Auckland, New Zealand Thuc-Quyen  Nguyen, MD Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA Itai M. Pessach, MD, PhD, MHA  The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Israel The Sacler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel Stephen  D.  Playfor, MBBS Department of Pediatric Critical Care, Royal Manchester Children’s Hospital, Manchester, UK Randy  P.  Prescilla, MD  Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston, MA, USA Jason Reynolds, MD  Pediatric Palliative Care, Cook Children’s Medical Center, Fort Worth, TX, USA Mark G. Roback, MD  Department of Pediatrics, University of Colorado School of Medicine, Children’s Hospital Colorado, Aurora, CO, USA James  A.  Roelofse, MBChB, MMed, PhD Department of Anesthesiology, University of Free State, Bloemfontein, South Africa University of the Western Cape, Cape Town, South Africa Stellenbosch University, Stellenbosch, South Africa University College London, London, UK Amber P. Rogers, MD, FAAP  Departments of Pediatrics and Anesthesiology, Perioperative and Pain Medicine, Texas Children’s Hospital, Houston, TX, USA Departments of Pediatrics and Anesthesiology, Baylor College of Medicine, Houston, TX, USA Joel S. Rosen, Esquire  Cohen, Placitella & Roth, P.C., Philadelphia, PA, USA Cyril  Sahyoun, MD Division of Pediatric Emergency Medicine, Children’s Hospital of Geneva, Hôpitaux Universitaires de Genève, Geneva, Switzerland Steven  M.  Selbst, MD Department of Pediatrics, Nemours/Alfred I. duPont Hospital for Children and Sidney, Kimmel Medical College at Thomas Jefferson University, Wilmington, DE, USA Neena  Seth, FRCA Evelina London Children’s Hospital, Guys and St Thomas’ NHS Foundation Trust, London, UK M.  Saif  Siddiqui, MD  Division of Pediatric Anesthesia and Pain Medicine, University of Arkansas for Medical Sciences and Arkansas Children’s Hospital, Little Rock, AR, USA Department of Anesthesiology, Arkansas Children’s Hospital, Little Rock, AR, USA Devona J. Slater, CHA, CHC, CMCP  Auditing for Compliance and Education, Inc. (ACE), Overland Park, KS, USA Sulpicio G. Soriano, MD  Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston, MA, USA Michael  J.  Stabile, MD, MBA Department of Anesthesia, Centennial Medical Center, Nashville, TN, USA St. Louis University School of Medicine, St. Louis, MO, USA

Contributors

Contributors

xix

Grant M. Stuart, MBChB, DCh, DA, FRCA  Department of Anaesthestics, Great Ormond Street Hospital NHS Trust, London, UK Paulo Sérgio Sucasas da Costa, MS, PhD  Federal University of Goias, Goiânia, Brazil The University of British Columbia, Vancouver, BC, Canada Joseph  D.  Tobias, MD Department of Anesthesiology & Pain Medicine, Nationwide Children’s Hospital, Columbus, OH, USA James  W.  Tom, DDS, MS, DADBA University of Southern California, Herman Ostrow School of Dentistry, Los Angeles, CA, USA Jacqueline  Cheuk  Kwun  Tse, MBBS, FHKCA Department of Anaesthesiology and Perioperative Medicine, Hong Kong Children’s Hospital, Hong Kong, China Gail A. Van Norman, MD  University of Washington Medical Center, Seattle, WA, USA Anesthesiology and Pain Medicine, Biomedical Ethics, University of Washington Medical Center, Seattle, WA, USA Robert A. Veselis, MD  Department of Anesthesiology and Critical Care Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA Pablo Osvaldo Sepúlveda, Dr. Med.  Hospital Base San Jose, Osorno, Chile Laszlo  Vutskits, MD, PhD Department of Anesthesiology, Pharmacology and Intensive Care, University Hospital of Geneva, Geneva, Switzerland Craig  S.  Webster, BSc, MSc, PhD Centre for Medical and Health Sciences Education, School of Medicine, University of Auckland, Auckland, New Zealand Graeme S. Wilson, MBChB, FCA(SA)  University of Cape Town, Red Cross War Memorial Children’s Hospital, Rondebosch, Western Cape, South Africa Stephen Wilson, DMD, MA, PhD  Department of Pediatric Dentistry, Nationwide Children’s Hospital, Columbus, OH, USA Blue Cloud Pediatric Surgery Centers, Glen Rock, PA, USA Bin Xue, MD  Department of Anaesthesiology, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China Ying  Xu, PhD, MD Department of Anaesthesiology, Children’s Hospital of Chongqing Medical University, Chongqing, China Lynne P. Yao, MD  Division of Pediatric and Maternal Health, Office of New Drugs, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, USA David A. Young, MD, MEd, MBA, FASA, CHSE  Baylor College of Medicine, Houston, TX, USA Texas Children’s Hospital, Houston, TX, USA Vivian  Man  Ying  Yuen, MD, FANCZA, FHKCA Department of Anaesthesiology and Perioperative Medicine, Hong Kong Children’s Hospital, Hong Kong, China

Part I Pediatric Sedation Outside the Operating Room

1

The History of Sedation Robert S. Holzman

Introduction The history of induced altered states as a means of tolerating the intolerable is as old as man and for eons has been alternately welcomed, worshipped, and vilified [1]. Ironically, as in ancient times, these three attitudes often coexist [2–4]. Is the history of sedation different from the history of anesthesia? They were and often continue to be inseparable, particularly for children,1 so we will focus on the various modalities and practices over time, emphasizing the differences but remaining in awe of the similarities through the ages. Most children in Ancient Egypt did not go to school. Instead boys learned farming or other trades from their fathers, and girls learned sewing, cooking, and other skills from their mothers. Some girls were also taught to read and write. Boys from wealthy families sometimes learned to be scribes. In Greece when a child was born, it was not regarded as a person until it was 5 days old when a special ceremony was held, and the child became part of the family. Parents were entitled, by law, to abandon newborn babies to die of exposure. Sometimes strangers would adopt abandoned babies. However, in that case, the baby became a slave. In Sparta children were treated very harshly. At the age of 7, boys were removed from their families and sent to live in barracks and treated severely to turn them into brave soldiers. They were deliberately kept short of food so they would have to steal  – teaching them stealth and cunning. Spartan girls learned athletics and dancing – so they would The Committee on Drugs of the American Academy of Pediatrics emphasizes that “the state and risks of deep sedation may be indistinguishable from those of general anesthesia.”2 The American Dental Association Council on Education defines general anesthesia to include deep sedation.3 The minimal distinction between deep sedation and general anesthesia has been recognized by the current author as well.4 1 

become fit and healthy mothers of more soldiers. Many of the inhabitants of Rome were slaves. Prisoners of war were made slaves, and any children slaves had were automatically slaves. The sons and daughters of well-to-do Romans went to a primary school called a ludus2 at the age of 7 to learn to read and write and do simple arithmetic. Girls left at the age of 12 or 13, and only boys went to secondary school where they would learn geometry, history, literature, and oratory (the art of public speaking). Infanticide was common and was a parent’s right if they didn’t want a child. The Spartans, for example, checked newborn infants for physical deformities and “mental” problems; if an abnormality was discovered, the child was tossed off a cliff. It was the state, not the father as in other Greek city-­ states, who determined whether a newborn male should live or die. Sparta was a military state and needed a good supply of robust babies. The difficulty with evaluating even relatively recent historical attitudes toward children is the paucity of documentation; this is particularly true with medical care. The common notion, introduced as a historical truth by Aries, was that children “were simply adults in miniature” [5]. Representations of children in medieval artwork depict them in adult clothing (Fig. 1.1). If they wore grown-up clothes, the theory goes, they must have been expected to behave like grown-ups. Similarly, literature rarely touched on the childhood years of its characters. The medical profession left nurses and mothers to deal with children. After all, because infants and small children were unable to speak and report their complaints, how could a learned doctor be expected to bother with them? These ideas, proposed with a dearth of primary evidence, likely devalued the love that ancient and medieval parents The same name applied to the school for gladiator training.

2 

R. S. Holzman (*) Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston, MA, USA e-mail: [email protected] © Springer Nature Switzerland AG 2021 K. P. Mason (ed.), Pediatric Sedation Outside of the Operating Room, https://doi.org/10.1007/978-3-030-58406-1_1

3

4

R. S. Holzman

Fig. 1.2 Ospedale degli Innocenti (Hospital of the Innocents). Florence, Italy. Commissioned in 1419; opened in 1445. Glazed blue terracotta medallions are mounted between the arches with reliefs of babies designed by Andrea della Robbia suggesting the function of the building. The insignia of the American Academy of Pediatrics is based on one of the medallions

Fig. 1.1  Children were “simply adults in miniature”

had for their children. While there certainly isn’t a great deal of medieval artwork that depicted children, the examples that survive do not universally display them in adult garb. Additionally, medieval laws existed to protect the rights of orphans. By the turn of the thirteenth century, English law recognized the dependency of the young and spoke of the needs they had for protection and support, especially around the time of the Black Death (1848–1850). In Bristol, as in London, officials made the guardianship of orphans a matter of public concern in order to protect inheritances. However, neither London nor Bristol protected the orphans of the poor and the propertyless. For example, in medieval London, the Orphan Court would place an orphaned child with someone who could not benefit from his or her death, and in this, the mayor had jurisdiction over the child’s financial and property assets. In keeping with the times, medieval medicine approached the treatment of children separately from adults. In general, children were recognized as vulnerable and in need of special protection. Early death, however, was an unfortunate but frequent event. Nonetheless, before the eighteenth century, few groups were more ignored medically than children. Death rates were high for a variety of reasons, mainly because of infectious diseases, but poor maternal care and prematurity led to difficulty caring for sick young babies. Nor was this restricted to the poor. Queen Anne, who ruled England from 1702 to 1714, had four miscarriages between 1684

and 1688, and not a single one of her 18 children survived to maturity. Such childhood mortality was considered natural, and parents viewed – and were encouraged to view – the loss of 75% of their infants before the age of 2 as a normal condition. Moreover, most doctors were not alarmed by the heavy death toll for babies, since they believed children to be outside of their scope of practice. When children received any treatment at all, they were drugged and bled just like their elders, because they were considered grownups in miniature. A growing humanitarian movement in the eighteenth century made the medical profession aware of its obligation to the underprivileged, including children. Thomas Coram (1668–1751), a bachelor, pitied the many babies abandoned on the highways leading to London. In 1739 he helped to found London’s Foundling Hospital. Though not a hospital in the modern sense, this child shelter drew generous aid from the medical profession and served to focus attention on proper childcare [6]. The first foundling hospital was established a millennium earlier by the archpriest of Milan in 787, although perhaps the most well-known of the foundling hospitals was the Ospedale degli Innocenti (Fig. 1.2). The most eloquent of the emerging pioneer “pediatricians” during the 1740’s educators was Dr. William Cadogan of London. He told mothers it was dangerous to swaddle babies tightly (in contrast to the swaddling portrayed in the medallions illustrated above), and his Essay Upon Nursing (1748) harshly criticized feeding infants an “unwholesome mess” (often, table scraps) (Fig.  1.3). Many mothers, he observed, were not nursing their babies “for fear of getting fat.” He also advocated frequent diaper changes (a source of disease) and gave infants a “right to kick” [7].

1  The History of Sedation

a

5

b

Fig. 1.3  Dr. William Cadogan, London pediatrician before the word “pediatrics.” Also illustrated is the title page of his Essay Upon Nursing and the Management of Children

There is a general perception that in the Middle Ages, children were not valued by their families or by society as a whole. On the other hand, it is important to remember that classical as well as medieval society was primarily an agrarian one. The family unit made the agrarian economy work. From an economic standpoint, nothing was more valuable to the working family than sons to help with the plowing and daughters to help with the household. To have children was, essentially, one of the primary reasons to marry. However, this hardly justifies a general perception of society’s indifference to childhood. Specific examples can be found, within the broad swath of cultural contexts over millenia, of kindness and progressively enlightened attitudes to childhood medical care. In the face of these facts, it is difficult to argue that people of ancient and medieval times were any less aware that children were their future than people are aware today that children are the future of the modern world. As a historical volte-face, it is intriguing to contemplate whether future history may reveal over-sentimentalization of children in our modern society.

I nebriation, Intoxication, Hallucination, and Anesthesia A Forme Fruste of the Sedation Continuum Alcohol is a fermentation product of many fruits and cereals, and its water solubility makes it a rapidly effective analgesic via glutamate inhibition and soporific via gamma-­ aminobutyric acid activation. Winemaking was first practiced in the Middle East about 6000–8000 years ago and was already well established in ancient Egypt. Although wine production was not well-developed in ancient Greece, wine was imported from other countries and often used for medicinal purposes. Benefitting from the breadth of their empire and diverse oenology exposure, the Romans developed the art of winemaking. Winemaking was ubiquitous in the ancient world – the Moors prepared date wines, the Japanese rice wines, the Indians (Mexico) made pulque from agave, the Vikings fermented honey to make mead, and the Incas made chi-

6

cha from maize. Modern beer making (yeast  – Saccharomyces cerevisiae) probably had its origin in Babylon as long ago as 5000–6000 bce. The addition of hops is a much more recent modification. Beer drinking and drunkenness were common in ancient Egyptian life; the Greeks learned their brewing skills from the Egyptians. Britons and Hiberni3 drank courni made from fermented barley. Wine remained an inebriant and intoxicant, however, until distillation technology was developed in the tenth century. Distillation exploits the fact that alcohol has a lower boiling point than water and therefore can be boiled out of an aqueous solution and condensed, approaching (but never achieving) purity—although 95% by volume is achievable. Liquors (such as rum or whisky) involve fermentation of sugarcane or barley, respectively. Liqueurs are usually produced by steeping fruits and/or herbs in brandy or vodka, with subsequent filtration to remove the vegetable residues. In this regard, absinthe, prepared from wormwood (Artemisia absinthium, A. maritime, or A. pontica), anise (Pimpinella anisum), and fennel (Foeniculum vulgare), plus nutmeg, juniper, and various other herbals, added to 85% alcohol, is then filtered and diluted to 75% alcohol by volume. Wormwood was the most important ingredient because of its psychotropic properties, recognized by ancient and medieval herbalists (Georg Ebers, Hippocrates, Dioscorides, John Gerard). The dose response of alcohol is interesting as a proxy for the continuum of sedation and general anesthesia. Mild intoxication occurs with a blood concentration of 30–50 mg/ dL (0.03–0.05%), and mild euphoria is achieved. Once the concentration has reached 100 mg/dL (0.10%), more serious neurological disturbances result in slurred speech and a staggering gait. At concentrations of 200 mg/dL (0.20%), vision and movement are impaired. Coma results at twice that concentration.

Ancient History Much of what we know in the twenty-first century about attempts to provide analgesia and sleep is derived from the written records of ancient civilizations in widely separated areas: China, India, Sumeria, and Egypt, for example. The recorded knowledge began approximately in the fourth millennium BCE, codifying oral drug lore that had undoubtedly preceded such codification by centuries. In rough chronological order of the records (but not by the use of the drugs themselves), we can begin with China.

Hibernia is the Latin name for Ireland; its people were the Hiberni.

3 

R. S. Holzman

Chinese Drug Lore The Pen Tsao (the symbols of which represent the compilation of medicinal herbs)4 was said to have been authored by Emperor Shen-nung in approximately 2700 bce. As the father of agriculture (the “Divine Husbandman”), he was said to have tasted all herbs in order to become familiar with their usefulness. Likewise, the Nei Ching was said to have been written by Emperor Hant-Ti (about 2700 bce). Although these texts describe the effects of naturally occurring herbs, the preparation of medicinals from herbs was attributed to I-Yin, a prime minister of the Shang dynasty (1767–1123 bce). The details of these preparations were recorded by making knots in strings, arranged vertically on a narrow bamboo surface. The ideograms utilized were uncannily similar to those chosen by Egyptian physicians in their hieroglyphs.5 As recording transitioned from string knots on bamboo to pen and paper, clinical cases and treatment recommendations were more easily recorded, initially by Chang Chung-Ching and the surgeon Hua Tuo (c. 140– 208), who probably used Cannabis indica (mafeisan6) for anesthesia (Fig.  1.4). This was probably no accident, as there is ample suggestion that Hua Tuo may have developed many of his medical ideas from Ayurvedic practices in an area of China richly influenced by Buddhist missionaries [8].

Hindu Drugs Brahman priests and scholars were the medical leaders in the earliest recorded histories, three of which are of foundational importance in Ayurvedic medicine: • Charaka Samhita (Compendium of Charaka) (second-­ century CE but copied from an earlier work) • Sushruta Samhita (Sushruta’s Compendium) (fifth-­ century CE) • Vagbhata (seventh-century CE) The Susruta detailed more than 700 medicinal plants, the most common of which were condiments such as sugar, cinnamon, pepper, and various other spices. Included among ben (pen; 本 “root”) and cao (tsao; 草 “herb”). The ideogram for “physician” (pronounced i) contained an arrow or a lancet in the upper half and a drug—or bleeding glass—in the lower half. 6  The name mafeisan combines ma (“cannabis; hemp; numbed”), fei (“boiling; bubbling”), and san (“break up; scatter; medicine in powder form”). Ma can mean “cannabis, hemp” and “numbed, tingling.” Other historians have postulated that mandrake or datura was used rather than cannabis, along with the wine. Still others have suggested hashish (bhang) or opium. 4  5 

1  The History of Sedation

7

Sumerian Drugs Agriculture developed in the area between the Tigris and Euphrates rivers (present-day Iraq), and a sophisticated cultivation of plant materials useful for the alleviation of symptomatic disease was not only practiced but also recorded. Nearly 30,000 clay tablets from the era of Ashurbanipal of Assyria (568–626 bce) were discovered in the mid-­nineteenth century near the site of Nineveh, capital of the neo-Assyrian Empire, with numerous references to plant remedies. Beers were especially well-developed in ancient Babylon. C. indica was known for producing intoxication, ecstasy, and hallucinations, especially when reinforced with hemp. This was all under the supervision of the priesthood. In addition, hallucinogenic mushrooms were employed in ancient Sumeria. Poppies were used mainly as a condiment in Sumerian life. Although there is no drug activity in the poppy leaves, fruit, or root, if the unripe seed capsule is opened, the white juice resulting from that is (raw) opium, the “latex” of which forms alkaloids as it dries. However, opium was not described (as far as we know) in the Ashurbanipal tablets.

Jewish Medicine

Fig. 1.4  Hua Tuo (c. 140–208 ce). The ancient texts Records of the Three Kingdoms and Book of the Later Han record Hua as the first person in China to use anesthesia during surgery, referring specifically to mafeisan. The illustration portrays Hua Tuo’s surgical and medicinal abilities as well as his use of moxibustion

them were descriptions of the depressant effects of Hyoscyamus (a source of scopolamine) and Cannabis indica. The eponymously named text (Susruta, c. 700–600 bce) described Susruta’s use of wine to the point of inebriation as well as fumitory cannabis in preparation for surgical procedures. Part of the difficulty with so many drugs was that they were not well codified and were prescribed in casual ways by numerous practitioners, who relied on (clinical) observation of effects [9].

Jewish medicine received significant contributions from the Babylonians during the Babylonian captivity (597–538 bce) as well as from the Egyptians during the Egyptian captivity (a date which is much less clear, based on 430 years of captivity prior to the Exodus, accepted as 1313 bce in rabbinic literature) [10]. Later Jewish medicine adopted Greek and Greco-Roman medical practices. Jewish potions were also prepared by the priesthood, the custodians of public health, for pain relief and the imparting of sleep during surgical procedures, venesection, and leeching. Rather than viewing sickness as divine retribution and its treatment ambivalent, physicians were regarded as the instrument through whom God could effect a cure. Jewish physicians therefore considered their vocation, whether curing illness or treating pain and suffering, as spiritually endowed. The use of opium as an analgesic and hypnotic drug was known, and warning was given against overdosing. “Sleeping dugs” (Samme de shinda) were probably hemp potions, but probably not opium derivatives [11].

Egyptian Medicine The major influence on the emerging Greek world of medicine came from Egypt. Our knowledge of their codification is relatively robust because of the medical papyri, most of which were hieratic (hieroglyphics or ideographs), compiled from around 2000–1200 bce (Table  1.1). These documents

R. S. Holzman

8 Table 1.1  List of Egyptian medical records Document Kahun Papyrus Edwin Smith Papyrus Ebers Medical Papyrus

Date 1900 bce 1600 bce

Hearst Medical Papyrus The Erman Document The London Papyrus The Berlin Papyrus The Chester Beatty Papyrus

1550 bce

1550 bce

1550 bce 1350 bce 1350 bce 1200 bce

Comment Primarily veterinary medicine Consists of 48 case histories; a well-organized surgical text Deals with medical rather than surgical conditions; emphasizes recipes Poorly organized; a practicing physician’s formulary Deals largely with childbirth and diseases of children Poorly organized; a practicing physician’s formulary Poorly organized; a practicing physician’s formulary Formulary for anal diseases; one case report

Fig. 1.5  Comparison of an opium poppy capsule and a base-ring juglet. An inverted opium poppy capsule on the left and a base-ring juglet from the Bronze Age (dated to Egypt’s 18th dynasty). Note that the solid pottery base ring takes the place of the serrated upper portion

were probably copied from older originals, as evidenced by the use of archaic terminology within the medical papyri, more characteristic of language from around 3000 bce. It is ironic, and somewhat puzzling, that despite the richness of ancient documentation from these artifacts, there is a paucity of information about narcotics and sedatives in ancient Egypt. Most of the suggestions about the use of such medications are by inference. For example, Ebers 782 cites shepnen of shepen (poppy seeds of poppy) to settle crying children. Interestingly, the poppy seed contains relatively little morphine; it is the latex produced from the incision of the seed pod that actually contains the active ingredient. Another suggestion, by inference, is that base-ring juglets were used to import opium from Cyprus in about 1500 bce, because of the resemblance of these juglets, when inverted, to a poppy head (Fig. 1.5) and the reported finding of mor-

of the capsule, but the flaring angle is almost identical. Overall, the outline of the body of the juglet almost parallels that of the poppy head, and its tall slender neck corresponds to the poppy’s thin stalk

1  The History of Sedation

phine in an Egyptian juglet from the tomb of Kha (nineteenth dynasty), although this has been disputed [12, 13]. Cannabis (C. sativa) was prescribed by the mouth, rectum, and vagina and delivered transdermally and by fumigation, yet central nervous system effects were not described. The London and Ebers papyri refer to mantraguru, an obvious common origin with mandrake or Mandragora. Some species of lotus (Nymphaea caerulea and N. lotus) are native to Egypt and contain several narcotic alkaloids that can be extracted in alcohol, leading to a logical hypothesis that lotus-containing wine might have additional narcotic effects. Ebers 209 and 479 both refer to preparations for the relief of right-sided abdominal pain and jaundice (respectively) containing lotus flower as an ingredient but directing that the lotus flower has to “spend the night” with wine and beer—conditions that would likely permit alkaloid extraction. It is therefore interesting that depictions of the lotus flower being sniffed are the only artifactual suggestion of the possible medical use of lotus (Fig. 1.6). Beer was thought to “gladden the heart” in general. Medicines mixed with beer—and combined with spells— were thought particularly effective. Beer and wine were also prescribed for children and nursing mothers. Childhood incontinence, for example, was treated in accordance with instructions from the Georg Ebers. The mother was to drink a cup of beer mixed with grass seeds and cyperus grass for 4 days while breastfeeding the child [14]. All over the world, indigenous people have learned the medicinal properties of plants in their environments. The remarkable accumulation of a fund of knowledge transmitted initially through oral tradition via specific lines of professional authority (physicians, priests, specialized castes of

9

drug-gatherers and preparers), and a sufficient amount of experience to provide the basis for a systematic analysis of efficacy undoubtedly took a long time. It is extraordinary, moreover, for its survival and consistency through the ages, laying the groundwork for classical civilization and beyond.

Classical History Greek Medicine Chaldo-Egyptian magic, lore, and medicine were transferred to the coasts of Crete and Greece by migrating Semitic Phoenicians and Jews. The stage was then set for incorporating ancient Egyptian drug lore into Greek medicine. Two prominent medical groups developed on the mainland of Asia Minor (the region located in the southwestern part of Asia comprising most of what is present-day Turkey): the group on Cnidos, which was the first, and then the group on Kos, of which Hippocrates (460–380 bce) was one member. While they were accomplished surgeons, they generally eschewed drugs, believing that most sick people get well regardless of treatment. Although Hippocrates did not gather his herbal remedies, he did prescribe plant drugs, and a cult of root diggers (rhizotomoi) developed, as did a group of drug merchants (pharmacopuloi). In Greece, plants were used not only for healing but also as a means of inducing death, either through suicide or execution; perhaps the best example was the death of Socrates.7 Later, Theophrastus (380–287 bc), a pupil of Aristotle (384–322 bce), classified plants and noted their medicinal properties. This was a departure from previous recordings, as Theophrastus analyzed remedies on the basis of their individual characteristics, rather than a codification of combinations as in Egyptian formularies. He provided the earliest reference in Greek literature to Mandragora [15]. The father of history, Herodotus (484–425 bce) (Fig. 1.7), left a detailed description of the mass inhalation of cannabis in the Scythian baths: The Scythians, as I said, take some of this hemp-seed, and, creeping under the felt coverings, throw it upon the red-hot stones; immediately it smokes, and gives out such a vapour as no Grecian vapour-bath can exceed; the Scyths, delighted, shout for joy, and this vapour serves them instead of a water-bath; for they never by any chance wash their bodies with water. [16]

Compression of the great vessels of the neck was also recognized as a form of inducing unconsciousness. It was recFig. 1.6  Stela (An upright stone slab or column typically bearing a commemorative inscription or relief design, often serving as a gravestone) of Ity (from the British Museum EA 586). Painted limestone Stela of Ity, dated to the 12th dynasty, c. 1942 bce. Ity’s many titles and the names of his mother, wife, sons, and daughters are listed. Note the illustration of the lotus flower being sniffed

As related by Plato in Phaedo, Socrates was sentenced to die by drinking poison hemlock (Conium maculatum). Coniline, the biotransformation product post-ingestion, has a chemical structure similar to nicotine, which exerts its inhibitory actions on nicotinic acetylcholine receptors resulting in neuromuscular blockade, respiratory failure, and death.

7 

10

R. S. Holzman

vera somno” (“poppies steeped in Lethe’s slumber”),9 while Ovid (43 bce–17/18 bce) also invoked the personification of Lethe by stating, “There are drugs which induce deep slumber, and steep the vanquished eyes in Lethean night.”10

Roman Medicine After the decline of the Greek empire following the death of Alexander the Great (323 bce), Greek medicine was widely disseminated throughout the Roman Empire by Greek physicians, who often were slaves. Dioscorides (c. 40–90 ce) described some 600 plants and non-plant materials including metals. His description of Mandragora is famous—the root of which he indicates may be made into a preparation that can be administered by various routes and will cause some degree of sleepiness and relief of pain [18]. Pliny the Elder (23–79 ce) described the anesthetic efficacy of Mandragora in the following manner: …(Mandragora is) given for injuries inflicted by serpents and before incisions or punctures are made in the body, in order to insure insensibility to pain. Indeed for this last purpose, for some persons the odor is quite sufficient to induce sleep. [19]

Fig. 1.7 Bust of Herodotus (484–425  bce). Roman copy of the Imperial Era (second-century CE) after a Greek bronze originally of the first half of the fourth-century BCE. Department of Greek and Roman Art, Gallery 162 Metropolitan Museum of Art, New York. (Photograph: Nguyen, PBM (2017, April 12). Herodotus. Ancient History Encyclopedia. Retrieved from https://www.ancient.eu/image/6501/. Published under a Creative Commons Attribution license: https://creativecommons.org/licenses/by/4.0/)

ognized that compression of the carotid8 arteries would result in unconsciousness and insensibility, as would pressure on the jugular veins. Aristotle acknowledged this saying of jugular vein compression, “if these veins are pressed externally, men, though not actually choked, become insensible, shut their eyes, and fall flat on the ground” [17]. The poets Virgil and Ovid described the soporific effects of opium. Virgil (70–19 bce) described the power of the poppy through the personification “Lethaeo perfusa papa-

The Greek word carotid means drowsiness, stupor, or soporific— hence the carotid artery is the artery of sleep. Galen incorporated its use as an adjective when he stated, “I abhor more than anybody carotic drugs.”

8 

In the first century, Scribonius Largus compiled Compositiones Medicorum and gave the first description of opium in Western medicine, describing the way the juice exudes from the unripe seed capsule and how it is gathered for use after it is dried. It was suggested by the author that it will be given in a water emulsion for the purpose of producing sleep and relieving pain [20]. Galen (129–199 ce), another Greek, in De Simplicibus (about 180 ad), described plant, animal, and mineral materials in a systematic and rational manner. His prescriptions suggested medicinal uses for opium and hyoscyamus, among others; his formulations became known as galenicals.

Islamic Medicine In 640 ce, the Saracens conquered Alexandria, Egypt’s seat of ancient Greek culture, and by 711 ce, they were patrons of learning, collecting medical knowledge along the way. Unlike the Christians, who believed that one must suffer as part of the cure, the Saracens tried to ease the discomfort of the sick. They flavored bitter drugs with orange peels and sweets, coated unpleasant pills with sugar, and studied the lore of Hippocrates and Galen. Persian physicians became the major medical teachers Virgil, Georgics 1. 78 As recorded in Fasti, a Roman calendar, 4:661

9 

10 

1  The History of Sedation

after the rise of the Baghdad Caliphate around 749 ce, with their teachings penetrating as far east as India and China. By 887  CE there was a medical training center with a hospital in Kairouan (present-day Tunisia) in Northern Africa. The most prominent of the Arab writers on medicine and pharmacy were Rhazes (865–925 ce) and Avicenna (930– 1036 ce), whose main work was A Canon on Medicine. The historical significance of this thread of recorded ancient medicine was that during the eleventh and twelfth centuries, this preserved knowledge was transmitted back to Christian Europe during the crusades. Avicenna noted the special analgesic and soporific properties of opium, henbane, and mandrake (Fig. 1.8) [21].

11

Medieval Medicine The first Christian early medieval reference to anesthesia is found in the fourth century in the writings of Hilary, the bishop of Poitiers [22]. In his treatise on the Trinity, Hilary distinguished between anesthesia due to disease and “intentional” anesthesia resulting from drugs. While St. Hilary does not describe the drugs that lulled the soul to sleep, at this time (and for the following few centuries) the emphasis remained on mandragora. From 500 to 1400 ce, the church was the dominant institution in all walks of life, and medicine, like other learned disciplines, survived in Western Europe between the seventh or eighth and eleventh centuries mainly in a clerical environment. However, monks did not copy or read medical books merely as an academic exercise; Cassiodorus (c. 485 ce–c. 585 ce), in his efforts to bring Greek learning to Latin readers and preserve sacred and secular texts, recommended books by Hippocrates, Galen, and Dioscorides while linking the purpose of medical reading with charity care and help. Conventional Greco-Roman drug tradition, organized and preserved by the Muslims, returned to Europe chiefly through Salerno, an important trade center on the southwest coast of Italy in the mid-900s. One of the more impressive practices documented at Salerno was intentional surgical anesthesia, described in Practica Chirurgiae in 1170 by the surgeon Roger Frugardi (Roger of Salerno, 1140–1195), in which he mentions a sponge soaked in “narcotics” and held to the patient’s nose. Hugh of Lucca (ca. 1160–1252) prepared such a sleeping sponge according to a prescription later described by Theodoric of Cervia (ca. 1205–1296). As an added precaution, Theodoric bound his patients prior to incision. The description of the soporific sponge of Theodoric survived through the Renaissance largely because of Guy de Chauliac’s (1300–1367) The Grand Surgery and the clinical practices of Hans von Gersdorff (c. 1519) and Giambattista della Porta (1535–1615), who used essentially the same formula of opium, unripe mulberry, hyoscyamus, hemlock, Mandragora, wood ivy, forest mulberry, seeds of lettuce, and water hemlock (Fig. 1.9).

Ether Fig. 1.8  Avicenna (930–1036 ce). “If it is desirable to get a person unconscious quickly, without his being harmed, add sweet-smelling moss or aloes-wood to the wine. If it is desirable to procure a deeply unconscious state, so as to enable the pain to be borne, which is involved in painful application to a member, place darnel-water into the wine, or administer fumitory opium, hyoscyamus (half dram dose of each); nutmeg, crude aloes-wood (four grains of each). Add this to the wine, and take as much as is necessary for the purpose. Or boil black hyoscyamus in water, with mandragora bark, until it becomes red, and then add this to the wine” [21]

Ether was discovered in 1275 ce by the Spanish chemist Raymundus Lullus (c. 1232–1315). This new discovery was given the name “sweet vitriol.” In 1540 ce, the synthesis of ether was described by the German scientist Valerius Cordus (1514–1544 ce) who carefully specified the materials to be used, the apparatus, and the procedure to be followed in order to distill “strong biting wine” (alcohol) with “sour oil of vitriol” (sulfuric acid). He recommended it for the relief of cough and pneumonia [25]. Paracelsus (1493–1541), a contemporary

12

Fig. 1.9  The alcohol sponge [23]. “Take of opium, of the juice of the unripe mulberry, of hyoscyamus, of the juice of hemlock, of the juice of the leaves of mandragora, of the juice of the wood-ivy, of the juice of the forest mulberry, of the seeds of lettuce, of the seeds of the dock, which has large round apples, and of the water hemlock—each an ounce; mix all these in a brazen vessel, and then place in it a new sponge; let the whole boil, as long as the sun lasts on the dog-days, until the sponge consumes it all, and it is boiled away in it. As oft as there shall be need of it, place this sponge in hot water for an hour, and let it be applied to the nostrils of him who is to be operated on, until he has fallen asleep, and so let the surgery be performed. This being finished, in order to awaken him, apply another sponge, dipped in vinegar, frequently to the nose, or throw the juice of the root of fenugreek into the nostrils; shortly he awakes” [24]

of Cordus, came surprisingly close to the recognition of ether as an anesthetic [26]. Later, in 1730, the German scientist W. G. Frobenius changed the name of sweet vitriol to ether.

Varied Preparations of Varying Potencies If the constituents of the plants were combined with fats or oils, they would penetrate the skin or could be easily absorbed via the sweat ducts in the axillae or body orifices such as the vagina or rectum. This would allow the psychoactive tropane

R. S. Holzman

Fig. 1.10  John Arderne (1307–1380). “An ointment with which if any man be anointed he shall suffer cutting in any part of his body without feeling or aching. Take the juice of henbane, mandragora, hemlock, lettuce, black and white poppy, and the seeds of all these aforesaid herbs, if they may be had, in equal quantities; of Theban poppies and of poppy meconium one or two drachms with sufficient lard. Braise them all together and thoroughly in a mortar and afterwards boil them well and let them cool. And if the ointment be not thick enough add a little white wax and then preserve it for use. And when you wish to use it anoint the forehead, the pulses, the temples, the armpits, the palms of the hands and the soles of the feet and immediately the patient will sleep so soundly that he will not feel any cutting” [27, 28]

alkaloids, especially hyoscine, access to the blood and brain without passage through the gut, thus avoiding the risk of poisoning. A few prominent surgeons offered statements about the mode of application of such salves or “ointments.” John Arderne (1307–1380) (Fig.  1.10), known for his success curing fistula-in-ano, and Andres De Laguna (1499– 1560) (Fig. 1.11), physician to Emperor Charles V and Philip II, provided unambiguous descriptions of soporifics.

1  The History of Sedation

Fig. 1.11  Andres de Laguna (1499–1560 ce). “…a pot full of a certain green ointment…with which they were anointing themselves…was composed of herbs…such as hemlock, nightshade, henbane, and mandrake…I had the wife of the public executioner anointed with it from head to foot…she…had completely lost power of sleep…no sooner did I anoint her than she opened her eyes, wide like a rabbit, and soon they looked like those of a cooked hare when she fell into such a profound sleep that I thought I should never be able to awake her…after a lapse of thirty-six hours, I restored her to her senses and sanity” [29]

The uncertainty of the potency and action of the narcotic drugs rendered their application dangerous, and by the end of the sixteenth century, such anesthetics had largely fallen into disrepute and disuse. Indeed, if physicians tried to use “narcotic” herbals in the middle of the seventeenth century, they were condemned, arrested, and fined or tried for practicing witchcraft [30]. Many of the early books were herbals, and Gerard (1545–1612) warned of the alkaloids “… this kind of Nightshade causeth sleepe…it bringeth such as have eaten thereof into a ded sleepe wherein many have died” [31].

The Scientific or Modern Epoch The divergence of herbalism (botany) and medicine began in the seventeenth century as part of the larger movement known alternatively as natural philosophy, scientific deism,

13

and the scientific revolution. An attempt to develop quantitative methodology characterized science, and at the forefront of these attempts was the chemical analysis of the active ingredients in medicinal plants. Following his clinical observation of poisoning in children who had mistaken water hemlock for parsnip root, Johann Jakob Wepfer (1620–1695) demonstrated dose-­ dependent toxic effects in dogs of the alkaloids eventually isolated as strychnine, nicotine, and conine [32, 33]. Thus, this early quantitative approach gave rise to the development of modern chemistry and pharmacology. This was first successfully applied to anesthetic pharmacology by Friedrich Wilhelm Adam Serturner (1783–1841) who, in 1805, described the isolation of meconic acid from the crude extract of opium and, in 1806, extracted opium. He further experimented with this crystal on dogs, finding that it caused sleep and indifference to pain and called this new substance morphine, in honor of the Greek god of dreams, Morpheus. This science of pharmacology—the interaction of chemistry with living matter—thus began to replace the ancient and descriptive materia medica of herbalism and set the stage for the advances of the second half of the nineteenth century, which included modern surgical anesthesia. The introduction of these drugs directly into the vascular system was developed by (Sir) Christopher Wren (1632– 1723) at Oxford in 1656 when he convinced his friend Robert Boyle (1627–1691) to experiment with a quill attached to a syringe through which opium was injected into a dog. What they found was that the opium made the dog stuporous, but did not kill him. Not long thereafter, in 1665, Johann Sigismund Elsholtz (1623–1688) administered opiates intravenously to humans in order to achieve unconsciousness, as described in his 1667 work Clysmatica Nova (Fig.  1.12) [34]. He performed early research into blood transfusions and infusion therapy and speculated that a husband with a “melancholic nature” could be revitalized by the blood of his “vibrant wife,” leading to a harmonious marriage. Direct transfusion of blood between animals was accomplished later that same year, and human transfusion followed 2 years later. Lamb’s blood was usually used, until James Blundell (1791–1878) transfused human blood into humans. By the 1830s, physiologists and elite doctors envisioned a level of unconscious life separable from the higher functions and the mind, including suffering. Advances in surgical thought, including more conservative and slower surgery, intensified the problem of pain for both patient and surgeon. By the mid-1840s, pain no longer seemed physiologically necessary or socially acceptable, but the intensive use of drugs known to diminish surgical pain was dangerous, and non-pharmacological alternatives such as mesmerism were highly contentious and controversial. Mesmerism, the predecessor of hypnosis, was based on Franz Anton Mesmer’s (1734–1815) belief that a magnetic

14

R. S. Holzman

Fig. 1.13  Mesmer practicing animal magnetism, from Hollander’s Die Karikatur und Satire in der Medizin, 1921. The title “Le Baquet de M. Mesmer” refers to Mesmer’s “tub” or cabinet, around which a group of patients would sit in order to press their afflicted body areas against the tub’s emerging metal rods. The patients would link their fingers to complete an “electric” circuit. The milieu was equally dramatic—an incense-filled room, haunting background music, mirrors, heavy drapes, and astrological symbols. There was a tremendous popular interest in medical applications of electricity. Coincidentally, Benjamin Franklin was the United States ambassador to France

Fig. 1.12  Illustration of venous injection, from Clysmatica Nova (1667). Note the disembodied hands delineating the vascular anatomy and illustrating the technique

field existed around people and could be controlled for health purposes to heal the sick. Mesmer’s strategy was to induce a trancelike state, rendering his patients hyperalert while asleep, a state referred to as “artificial somnambulism” by the Marquis de Puysegur (1751–1825), which eventually became known as “hypnosis” (de Cuvillers, in 1820) (Fig. 1.13) [35]. Hypnosis was used as an adjunct to surgery in the 1830s by Cloquet (mastectomy) and Elliotson, and ironically (in 1846) Esdaile (1808–1859) reported on the use of hypnoanesthesia in approximately 300 surgical patients in India [36]. Because the public demonstration of ether was virtually simultaneous, medical applications of hypnosis rapidly fell into disuse, and, intriguingly, it was relegated to entertainment—much like nitrous oxide before the “acceptance” of chemically induced anesthesia. Turnabout was fair play. Hypnosis is making a comeback for sedation, especially with children [37–39], and has been shown to reduce

required amounts of propofol and lidocaine, with accompanying reductions in pain, nausea, fatigue, discomfort, and emotional upset. It has also been shown to reduce the cost per patient by more than $750, mainly due to a shorter time in the operating room [40, 41]. The time was thus ripe for the integration of science and medicine and the introduction of pneumatic medicine by Thomas Beddoes (1803–1849). He was committed to the notion that chemistry, especially the use of medicinal gases, could transform medicine and was convinced that the newly discovered respirable gases, nitrogen, hydrogen, and oxygen, could be therapeutic for various lung conditions such as tuberculosis [42]. It was his employee Humphrey Davy’s experiments with nitrous oxide that fueled the therapeutic use of gases, including his experiments with nitrous oxide’s ability to be breathed longer than any of his other experimental gases (except air and oxygen) with animals showing an initial period of excitement, followed by exhaustion. Furthermore, he noted that even if the animal stopped breathing gas before complete exhaustion was reached, it was still possible to restore “healthy living action” by letting the animal breathe atmospheric air. The “peculiar changes” in the blood and organs were therefore reversible. This concept of death as a process, a continuum, rather than an absolute, was evolutionary and revolutionary at the time yet ironically reminiscent of the continuum of life and death of classical civilization!

1  The History of Sedation

Henry Hill Hickman (1800–1830) was born in the year Humphrey Davy suggested that nitrous oxide might be used for pain relief during surgery. Hickman, a country doctor, conceived, promoted, and attempted to practice “pain-free surgery.” Hickman experimented at a time when understandings of asphyxia were changing and death began to be conceived of as a process. Medical research began to focus on resuscitation and the various techniques that could restore life in a body lacking a pulse or respiration. Thus Hickman understood suspended animation as a form of asphyxia, a state in which respiration had been suspended but life still existed—hence he use bellows during a 17-min amputation of the leg of a dog. It is clear too that Hickman was incorporating a new understanding of the nervous system from the work of Charles Bell (1774–1842) in Britain and François Magendie (1783–1855) in France in the 1810s, supporting the separation of mind and the body. Hickman based his experiments on the belief that if applied to humans, the key benefit would be the suspension of the mind of the patient and thus the absence of anticipation of suffering, as well as the relief of physical pain. Hickman advocated what he called “suspended animation” (general anesthesia) for surgery on humans as well. He had the right idea about inhalation anesthesia but unfortunately, in selecting carbon dioxide, picked the wrong agent. Carbon dioxide can indeed induce unconsciousness, but the gas also often results in panic attacks. In larger quantities, it is lethal. In a scathing letter to the editor of The Lancet in 1826, Hickman’s work was brutally criticized in an article entitled “Surgical Humbug” [43]. In his attempt to seek support abroad, Hickman decided to try his luck in Paris in 1828 and presented a paper to King Charles X.  The paper was forwarded to the Academie Royale de Medecine. A committee was set up to investigate Hickman’s proposal for painless surgical operations on humans but was unsupported by French scientists. He went back to England to live out his remaining years and work hard in his poor practice, dying of tuberculosis 2 years later at 30 years of age. Interest in the intravenous methods persisted as well, and Pierre-Cyprien Oré injected chloral hydrate in 1872 in order to produce an anesthetic state in humans (following animal experimentation). Again, unfortunately, an incorrect drug was chosen, because intravenous chloral hydrate has a very narrow therapeutic margin. Emil Fischer (1852–1919) synthesized barbital in 1902, and although it was ineffective as an intravenous anesthetic because its onset and termination was too slow, hexobarbital (Evipal) followed 30 years later and was first reported for anesthetic use in 1932. Sodium thiopental followed in 1943.

The Modern Story of Anesthesia The modern story of anesthesia began with the reaction in Philadelphia to Humphry Davy’s (1778–1829) account of nitrous oxide and its biological effects. In 1808, William

15

P. C. Barton (1786–1856) emphasized the brain disorientation caused by inhaling nitrous oxide and cited Davy. Meanwhile, an anonymous note, often ascribed to Michael Faraday, indicated that the inhalation of ether would produce effects similar to those of nitrous oxide. In 1839, a medical student, William E. Clarke (1818– 1878) in Rochester, New York, began the fad of ether frolics among young people. He is said to have given ether for extraction of a tooth in 1842. In Jefferson, Georgia, Crawford W. Long (1815–1878) noted that one of the participants in an ether frolic fell heavily but seemed to lack pain. On March 30, 1842, Long gave ether by inhalation to a patient for removal of a neck tumor; there was no evidence of pain. Modern pediatric surgical anesthesia also began with Long, who administered ether anesthesia to an 8-year-old boy named “Jack” for a toe amputation on July 3, 1842. The boy had no evidence of response to the amputation and reported no pain or recall of the procedure [44]. Many children had surgery without the benefit of anesthesia until that time, as they “did not seem to appreciate pain and would seem insensible even during the operation.” Others thought that young children lacked the intelligence to suffer and were therefore ideal candidates for surgery in any event [45]. Unfortunately, he failed to report his anesthetic success for several years. William T. G. Morton (1819–1868), a student at Harvard Medical School, learned of sulfuric ether and practiced anesthetizing various small animals at his home. He tried to perfect an inhaling device, and a demonstration was arranged at the Massachusetts General Hospital on October 16, 1846, a turning point in the history of medicine. Gardner Quincy Colton (1814–1898) first gave nitrous oxide for anesthetic purposes to Horace Wells in 1844 and revived its use in dentistry for dental extractions in 1863. Colton and Smith established the Colton Dental Institute in New York in 1864 and over a period of 30 years treated 186,500 patients without “a single accident from the gas” [46]. In England, Alfred Coleman (1828–1902) became the chief advocate for the use of nitrous oxide in dentistry. Clinical administration, of course, was not without its risks. In the latter half of the nineteenth century, before co-administration with oxygen, 100% nitrous oxide was administered, in the sitting position (with the head flexed in order to prevent the tongue from falling backward onto the hard palate): …Until breathing was rapid, the face was (at first) pale, then cyanotic…The aspect of the patient is at this time ghastly in the extreme, there being every physical indication of impending asphyxia…These appearances are coincident with anesthesia sufficiently profound for the needs of minor surgery and the inhaler must be withdrawn and the operation swiftly performed. [47]

In stark contrast to the divergence of the medical care of children from adults before the mid-nineteenth century, with the beginning of the modern anesthetic era, great advances

16

R. S. Holzman

were provided to all surgical patients. Pediatric patients were involved early in all forms of anesthetic administration because of their suitability, basic health, and in an effort to make more humane the physiologic and emotional ordeal of the surgical experience. Pediatric reports followed quickly after the Ether Dome demonstration at the Massachusetts General Hospital in 1846; in the same year, an 8-year-old patient of Dr. James Robinson reported that he had been “in heaven or somewhere else” during an ether anesthetic for dental surgery [48]. John Snow, the first full-time physician anesthesiologist, insightfully observed in the same year that “children are, indeed, amongst the most favourable subjects for ether, recovering from its effects as promptly as they are brought under its influence...” [49]. As chloroform evolved into the principal British inhalation anesthetic in the late nineteenth and early twentieth century, it is the work of Snow that further elaborates the now better understood but hardly more elegantly described findings of inhalation pharmacokinetics: The effects of chloroform are more quickly produced and also subside more quickly in children than adults, owing no doubt to the quicker breathing and circulation, etc. [50]

Snow reported enthusiastically on his experience with a series of 186 babies less than 1 year of age and noted: There have been no ill effects from it (chloroform) either in these cases, or in those of chil-dren more advanced in life; it is worthy of remark that none of the accidents from chloro-form which have been recorded, have occurred to young children. [50]

Along with the recognition of the suitability of anesthetics for surgical procedures in children, and the popularity and relative ease of administration, came the inevitable recognition of accompanying risks. The first recorded anesthetic death—a healthy 15-year-old girl named Hannah Greener, undergoing toenail excision under chloroform anesthesia—occurred in 1848, within 2 years of the Ether Dome demonstration. Deaths from cardiac arrest and hepatic failure were reported throughout the second half of the nineteenth century, while chloroform’s popularity prevented it from being replaced with ether as a safer agent. Frequently, these deaths were in young people, right at the beginning of the anesthetic, when the patient was lightly anesthetized and nervous. It was only at the beginning of the twentieth century that it became clear that the interaction of high levels of endogenous catecholamines along with light levels of chloroform anesthesia could result in ventricular fibrillation. Occasionally, mixtures of chloroform and ether were utilized to decrease the risk of circulatory instability and ectopy [51]. Bodington’s observation about the contribution of oxygenation to outcome, made early in the history of anesthesia (1873), was prescient:

From what I have read my strong impression is that the occasional bad results are not due positively to the anesthetic itself, but negatively to the sudden absence of oxygen occurring inadvertently in the course of administration of the agent, notwithstanding the ingenious methods employed. [52]

Supplemental oxygen was introduced by Hewitt at the turn of the century [53]. The presumptive risks of hypoxia associated with nitrous oxide (especially the technique of “secondary saturation” practiced by clinicians since the late nineteenth century) [54] were finally proven by C.  B. Courville (a neuropathologist) in 1939 [55], although the concepts did not enter into clinical practice until after World War II. The principal advance, however, was the reformulation of the goals of nitrous oxide administration—for its sedative rather than anesthetic or analgesic properties. With patients never reaching the excitement stage, nitrous oxide was used to produce sedation and local anesthesia to control pain. The formation of the American Dental Society of Anesthesiology in 1953 furthered this concept. There were additional “sleep-producing” agents available in the second half of the nineteenth century. For example, it was recognized by Robert Glover that potassium bromide would cause drowsiness in animals and by Charles Locock that it would effectively treat epileptic seizures in obstetrical patients being treated for dysmenorrhea. Behrend reported its use for the treatment of insomnia, nervous excitement, and irritability. This led to the therapeutic use of “bromides” (of lithium, sodium, and potassium) as anticonvulsants. It was only a short time later that chloral hydrate was introduced by Liebreich as a soporific for medical purposes [56] as well as more nefarious purposes (it was the chief ingredient in the “Mickey Finn” cocktail, for which the bartender, Michael Finn, was tried in 1903  in Chicago). Additional soporifics were paraldehyde, ethanol, sulfonal, diethyl-­ malonyl urea (veronal or barbital), and phenyl-ethyl malonyl urea (luminal or phenobarbital).

“Modern” Sedation and Analgesia Services There is an inseparable continuum, particularly in pediatrics, between general anesthesia and sedation and analgesia. Not surprisingly, it was the early efforts of dental surgeons at the beginning of the twentieth century that spearheaded ambulatory anesthesia, much as early general anesthesia was associated with dental procedures. Many dentists produced, purified, and stored their own nitrous oxide. The first-day surgery began at the Royal Hospital for Sick Children in Glasgow with pediatric surgeon James Nicholl, who began to operate on children as outpatients. In 1909 he reported a 10-year history of almost 9000 operations on children as outpatients; unfortunately, there is no mention of anesthesia [57]. In 1916, Ralph Waters (1883–1979) opened the

1  The History of Sedation

Downtown Anesthesia Clinic in Sioux City, Iowa, caring for dental and minor surgery patients but avoiding ether in favor of nitrous oxide along with the selection of appropriately short surgical procedures such as dental extractions, circumcisions, simple fractures, or incision and drainage of abscesses [58]. Intermittently, pediatric anesthesiologists filled the role of sedation experts in order for children to tolerate unpleasant diagnostic procedures (Fig. 1.14). “Twilight sleep” was also introduced in the early part of the twentieth century; it is a term that persists to this day, perhaps because of its colorful name, which originated from the German Dammerschlaf, introduced by Gauss in 1906 to describe the state of clouded consciousness produced by a combination of scopolamine and morphine. The technique had actually been introduced several years earlier, but Gauss (and obstetrician Bernhard Kronig) broadened its use in hundreds of patients at the Frauenklinik of the State University of Freiburg and reported their results in 500 patients [59]. The impact, particularly among women, in the early twentieth century was astounding—reporters from the Ladies’ Home Journal, the Woman’s Home Companion, and McClure’s journeyed to Germany to investigate the heralding of a new age in obstetrical analgesia. Popular favorable reports rapidly followed, galvanizing a political movement for obstetrical pain relief largely advanced by women. In a rallying statement, the Ladies’ Home Journal correspondent who eventually wrote Truth About Twilight Sleep stated: I now make my last appeal to every woman who has read this book to take up the battle for painless childbirth where I left off…Fight not only for yourself, but for your sisters, your sex, the cradle of the human race…Through Twilight Sleep a new era has dawned for woman and through her for the whole human race. [60]

17

The technique was not perfect. There was wide variation in the response to the drug combination, from incomplete analgesia to incomplete erasure of memory. Patients continued to groan and scream in agony; they just could not remember afterwards. Kronig would therefore not allow the presence of any family members—nor reporters or professional observers—to directly verify the efficacy of the technique. The end result, however, was that the majority of patients would not recall anything about the birth and would awaken after delivery and state that they hoped the labor would begin soon, which then gave rise to the debate about whether there is pain if there is no memory of pain. It further highlighted a problem that Gauss faced every day—his attempts to standardize the dose were difficult at best. He expressed it clearly: “If you could trust to having an average women, you could use an average dose; but the dose is easier to standardize than the woman.” Competing institutions adopted Gauss’ recommendations, with results ranging from praise to condemnation. In America, similar ambivalence was encountered. Twilight sleep was adopted wholeheartedly and enthusiastically at Long Island College Hospital in Brooklyn (by patient request) but was abandoned at Johns Hopkins. This controversy reflected the narrow therapeutic range of the technique, again summarized succinctly by Gauss: “Twilight Sleep is a narcotic condition of extremely narrow breadth, like a narrow mountain crest. To the left of it lie the dangers of too deep action, with narcosis and absence of birthpains; to the right, the danger of shallow action, with retention of consciousness and sensibility of pain.” The tensions developed between the medical profession, the medical press, and the public are outlined very thoughtfully by Caton in What a Blessing She Had Chloroform (1999). Waters’ prescience was followed by a long gap, until the 1960s, when increasing interest in employing shorter-acting anesthetic strategies with more rapid return to “street fitness” predated the explosion onto the medical diagnostic scene of computed tomography (1974), magnetic resonance imaging (1977), interventional radiology procedures, cardiac catheterization (diagnostic and interventional), and various other imaging modalities. In addition, further miniaturization and engineering improvements continued for both gastrointestinal and pulmonary endoscopy and the use of radiation therapy as an adjunct to surgical and medical treatment of cancer patients. All of these took place in nontraditional anesthetizing locations, popularly known as “outfield” anesthesia.

The Future of Sedation Fig. 1.14  A cachectic child undergoing intrapulmonary contrast injection via an intratracheal catheter for radiographic evaluation of tuberculosis. The tracheobronchial tree was topically anesthetized with local anesthetic, and intermittent sedation was provided by the inhalation of nitrous oxide. (From a pediatric anesthesia training film made by Dr. M. Digby-Leigh in 1947)

As an increasing number of procedures are developed that are accessible by percutaneous, intravascular, or natural orifice routes, they will be less painful in both the awake and asleep state. However, the need for motionlessness for chil-

18

dren as well as adults will remain, especially as these imaging techniques and procedures are likely to be longer and require increasingly sophisticated instrumentation. At the same time, progress will inevitably continue in understanding the neurophysiology of pain mechanisms as well as consciousness. With every scientific and clinical development since the beginning of surgical anesthesia, creative individuals were able to adapt to the challenges of caring for children. This wonderful history is a part of the legacy of anesthesia and its contribution to the humane and safe care of children.

References 1. Holzman R. Uncertainty by choice: anesthesia and the children of night. J Clin Anesth. 2002;14(1):46–51. 2. Committee on Drugs American Academy of Pediatrics. Guidelines for monitoring and management of pediatric patients during and after sedation for diagnostic and therapeutic procedures. Pediatrics. 1992;89(6):29–34. 3. Holzman R, Cullen D, Eichhorn J, Philip J. Guidelines for sedation by nonanesthesiologists during diagnostic and therapeutic procedures. J Clin Anesth. 1994;6:265–76. 4. Kallar S, Dunwiddie W. Conscious sedation. In: Wetchler B, editor. Outpatient anesthesia. Problems in anesthesia, vol. 1. Philadelphia: Lippincott; 1988. p. 93–100. 5. Aries P.  Centuries of childhood: a social history of family life. New York: Knopf; 1962. 6. McClure R. Coram’s children: the London foundling hospital in the eighteenth century. New Haven: Yale; 1981. 7. Wilson A. Ritual and conflict: the social relations of childbirth in early modern England. Farnham: Ashgate; 2013. 8. Mair V.  The biography of Hua-t’o. In: Mair V, editor. History of the three kingdoms. The Columbia anthology of traditional Chinese literature. New York: Columbia University Press; 1994. p. 688–96. 9. Chopra R. Indigenous drugs of India: their medical and economic aspects. Calcutta: Art Press; 1933. 10. Finegan J.  Handbook of biblical chronology: principles of time reckoning in the ancient world and problems of chronology in the Bible. Peabody: Hendrickson Publishers; 1998. 11. Friedenwald H.  The Jews and medicine; essays by Harry Friedenwald. Baltimore: The Johns Hopkins Press; 1944. 12. Merrillees R.  Opium trade in the bronze age levant. Antiquity. 1962;36(144):287–92. 13. Nunn J.  Ancient Egyptian medicine. London: University of Oklahoma Press; 1996. 14. Mark J. Egyptian medical treatments. Ancient history Encyclopedia. Surrey: Ancient History Encyclopedia; 2017. 15. Holzman R. The legacy of Atropos, the fate who cut the thread of life. Anesthesiology. 1998;89(1):241–9. 16. Herodotus. The histories. New  York: Everyman’s Library; 1997; Knopf; Random House; 440 BCE. 17. Aristotle. The history of animals [Internet]. 1907. Available from: http://classics.mit.edu//Aristotle/history_anim.html. 18. Anazarbus Dioscorides of Pedanius. The Greek herbal of Dioscorides, illustrated by a Byzantine A.D. 512 Englished by John Goodyear A.D. 1655. Oxford: Oxford University Press; 1933. 19. Pliony the Elder. The natural history (c. 77 AD). London: Taylor and Francis; 1855. 20. Leake C. An historical account of pharmacology to the 20th century. Springfield: Charles C. Thomas; 1975.

R. S. Holzman 21. Avicenna. Qanun dar tibb (Canon of medicine). Tihran: Surush; 1987. 22. Humphrey J, Oleson J, Sherwood A. Greek and Roman technology: a sourcebook: annotated translations of Greek and Latin texts and documents. London: Routledge; 1998. 623 p. 23. Schilling D.  Diebold Schillings Spiezer Bilderchronik: kommentar zur Faksimile-Ausgabe der Handschrift Mss. hist. helv. I. 16 der Burgerbibliothek Bern / herausgegeben von Hans Haeberli und Christoph von Steiger; mit Beiträgen von Vinzenz Bartlome. Luzern: Faksimile Verlag; 1513. 24. Borgognoni T.  The surgery of Theodoric ... Tr. from the Latin. New York: Appleton-Century; 1955. 25. Cordus V. Annotationes in Pedacii Dioscordies Anazarbei de materia medica. Strasbourg/Argentorati: Excudebat Iosias Rihelius; 1561. 26. Temkin O, Rosen G, Zilboorg G, Sigerist H.  Defensiones, in four treatises of Theophrastus von Hohenheim called Paracelsus. Boston: Johns Hopkins Press; 1941. 27. Fraenkel W.  Uber Narkosen und Weckmittel des John Arderne, Schmerz-Narkose-Anesthesie. Leipzig. 1937:74–8. 28. Piomelli D, Pollio A.  A study in Renaissance psychotropic plant ointments. Pubblicazioni dell aStazione Zoologica di Napoli Section Ii: Hist Phil Life Sci. 1994;16:241–73. 29. Rothman T.  De Laguna’s commentaries on hallucinogenic drugs and witchcraft in Dioscorides’ Materia Medica. Bull Hist Med. 1972;46:562–7. 30. Summers M. The history of Wtchcraft and demonology. New York: Knopf; 1926. 31. Gerard J. The herball, or generall historie of plantes. London: Adam Islip, Joice Norton and Richard Whitakers; 1633. 32. Eichenberger P. Autobiographic material on Johann Jakob Wepfer (1620–1695) in correspondence with Johann Conrad Brotbeck (1620–1677). Gesnerus. 1967;24:1–23. 33. Maehle A. Scientific and moral justification of toxicologic animal trials in the 17th century. Johann Jakob Wepfer and Johann Jakob Harder. Gesnerus. 1986;43:213–21. 34. Gladstone E. Clysmatic Nova (1665): Elsholtz’ neglected work on intravenous injection. Cal West Med. 1933;39(2):119–23. 35. Holländer E.  Die Karikatur und Satire in der Medizin: mediko-­ kunsthistorisches Studie. Stuttgart: Verlag von Ferdinand Enke; 1921. 36. Esdaile J. Mesmerism in India, and its practical application in surgery and medicine. New York: Arno Press; 1976. 37. Butler L, Symons B, Henderson S, Shortliffe L, Spiegel D. Hypnosis reduces distress and duration of an invasive medical procedure for children. Pediatrics. 2005;115:e77–85. 38. Calipel S, Lucas-Polomeni M, Wodey E, Ecoffey C. Premedication in children: hypnosis versus midazolam. Pediatr Anesth. 2005;15:275–81. 39. Saadat H, Drummond-Lewis J, Maranets I, Kaplan D, Saadat A, Wang S, et  al. Hypnosis reduces preoperative anxiety in adult patients. Anesth Analg. 2006;102:1394–6. 40. Montgomery G, Bovbjerg D, Schnur J, David D, Goldfarb A, Weltz C, et al. A randomized clinical trial of a brief hypnosis intervention to control side effects in breast surgery patients. J Natl Cancer Inst. 2007;99:1304–12. 41. Spiegel D. The mind prepared: hypnosis in surgery. J Natl Cancer Inst. 2007;99(17):1280–1. 42. Snow S.  Operations without pain: the practice and science of anaesthesia. In: Pickstone J, editor. Victorian Britain. Hampshire: Palgrave Macmillan; 2006. 43. Antiquack. Surgical Humbug. Lancet. 1826;5(127):646–7. 44. Pierson A. Early operation for harelip. Am J Med Sci. 1852;24:576. 45. Bigelow H. Etherization. Trans AMA. 1848;1:211. 46. Mead S. Anesthesia in dental surgery. St. Louis: C.V. Mosby Co.; 1935.

1  The History of Sedation 47. Litch W.  Anaesthesia and anaesthetics. In: Litch W, editor. The American system of dentistry, vol. 3. Philadelphia: Lea Brothers; 1887. p. 144–98. 48. Robinson J. A treatise on the inhalation of the vapour of ether for the prevention of pain in surgical operations. London: Webster & Co; 1847. 16 p. 49. Snow J. On the inhalation of the vapour of ether. London: Churchill; 1848. 28 p. 50. Snow J.  On chloroform and other anaesthetics, vol. 49. London: Churchill; 1858. p. 258–9. 51. Flagg P. The art of anaesthesia. 4th ed. Philadelphia: JB Lippincott; 1916. 52. Bodington G. Anaesthetics. Lancet. 1873;i:32. 53. Smith W. A history of nitrous oxide and oxygen anaesthesia. XI: the use of Hewitt’s “mixture” and the development of nasal administration. Br J Anaesth. 1972;44:106–14.

19 54. McKesson E.  Primary and secondary nitrous oxide saturation for relaxation and as a test of the patient’s capacity for operation. Can Med Assoc J. 1921;11(2):130–6. 55. Courville C. Untoward effects of nitrous oxide anesthesia. Mountain View: Pacific Press Publishing Association; 1939. 56. Liebreich O.  Das Chloralhydrat: Ein neue Hypnoticum und Anaestheticum. Arch Deutsch Ges Psychiat. 1869;16:237. 57. Nicoll J. The surgery of infancy. Br Med J. 1909;2(2542):753–4. 58. Waters R.  The downtown anesthesia clinic. Am J Surg (Anesth Suppl). 1919;33:71–3. 59. Gauss C. Births in Artificial Dammerschlaf. Translated from Archiv Für Gynäkologie. 1906;78:579–631. Berlin 1914. 60. Rion H. The truth about twilight sleep. New York: McBride, Nast & Co.; 1915.

2

Sedation Policies, Recommendations, and Guidelines Across the Specialties and Continents Joseph P. Cravero

Introduction The practice of pediatric sedation involves a wide variety of pediatric surgical and medical sub-specialists. There are no “universally” applicable and acceptable guidelines that apply to all the physicians and nurses who are take part in sedating children for procedures. A number of guidelines, policies, and recommendations for sedation care have been promulgated by different subspecialty societies over the last 40  years. This chapter will consider development of these guidelines and put them into context and perspective. There are several forms of guidelines – those that come in the form of “statements,” “practice advisories,” “clinical policies,” clinical practice guidelines,” or “recommendations.” These documents range from those that contain general descriptions of appropriate monitoring and treatment to those that offer very specific guidelines on the use of particular drugs or pre-sedation nil per os (NPO) intervals. There is variability in the recommendations that pediatric subspecialty societies make concerning the specifics of sedation care, but the most important elements are remarkably similar. It should also be noted that the methodologies used to produce these guidelines vary from organization to organization. For example, in the American Academy of Pediatrics (AAP), guidelines are put together by a workgroup on sedation from the Committee on Drugs [1–3].While these guidelines are based on a careful consideration of the available literature and review by a number of experts in pediatric specialties, the exact nature of how studies were “weighted” and conclusions are drawn is not explicitly described. On the other hand, the most recent guidelines from the British National Institute for Healthcare and Clinical Excellence (see International Guidelines section), American Society of J. P. Cravero (*) Harvard Medical School, Boston, MA, USA Department of Anesthesiology, Critical Care, and Pain Medicine, Boston Children’s Hospital, Boston, MA, USA e-mail: [email protected]

Anesthesiologists [3] (ASA), and American College of Emergency Physicians (ACEP) [4] are founded on a structured, evidence-based, review of pediatric sedation literature, and the methodologies are explicit. Even in publications such as this, the absence of high-quality controlled trials in pediatric sedation necessitates that many of the most important aspects of these guidelines are based on “consensus” (or some interpretation of the data) rather than strictly on the evidence. This chapter will review a number of sedation guidelines from organizations centered in the United States and internationally. Some are published in peer-reviewed literature, and some are available on society websites. As the comparison of guidelines can be quite subtle, and the acronyms for various organizations are incredibly confusing, the various guidelines in this chapter will be organized by their sponsoring organization.

 merican Academy of Pediatrics (AAP) A Guidelines “The AAP Guidelines for Monitoring and Management of Pediatric Patients During and After Sedation for Diagnostic and Therapeutic Procedures” are the most widely cited, and applied, guidelines with respect to pediatric sedation. While other statements from the AAP have expanded on the importance of the use of sedation and analgesia for children [5–7], the sedation guidelines specifically address the clinical provision of sedation for children. They have influenced the creation of safe sedation systems across the USA and internationally. Much of their lexicon and recommendations have been largely adopted by The Joint Commission (TJC) and by regulatory bodies in Europe and Australasia in evaluating institutional compliance for safe sedation standards. The first AAP guideline for pediatric sedation was written in response to three dental deaths in 1983 (published in 1985) [8] on behalf of the American Academy of Pediatrics (AAP) Section on Anesthesiology. Written in collaboration with the American Academy of Pediatric Dentistry (AAPD) and the

© Springer Nature Switzerland AG 2021 K. P. Mason (ed.), Pediatric Sedation Outside of the Operating Room, https://doi.org/10.1007/978-3-030-58406-1_2

21

22

American Society of Anesthesiologists (ASA), the purpose was to develop a framework from which improved safety could be developed for children requiring sedation in order to perform a needed procedure. This initial guideline emphasized standardization on issues such as the need for informed consent, appropriate fasting prior to sedation, frequent measurement and charting of vital signs, the availability of age and size appropriate equipment, the use of physiologic monitoring, the need for basic life support skills, and proper recovery and discharge procedures. The concept of an independent observer whose only responsibility is to monitor the patient was introduced for deeply sedated pediatric patients. Advanced airway and resuscitation skills were encouraged but not specifically required for deep sedation providers. These original guidelines defined three terms for depth of sedation: conscious sedation, deep sedation, and general anesthesia. The descriptive term “conscious sedation” was defined as “A medically controlled state of depressed consciousness that allows the protective reflexes to be maintained; retains the patient’s ability to maintain a patent airway independently and continuously; and permits an appropriate response by the patient to physical stimulation or verbal command, e.g. ‘open your eyes’.” In 1992 the Committee on Drugs of the AAP revised the 1985 guideline [1]. This new iteration recognized that a patient could readily progress from one level of sedation to another and that the practitioner should be prepared to increase vigilance and monitoring as indicated. Pulse oximetry was recommended for all patients undergoing sedation. This new guideline also discouraged the practice of administering sedation at home by parents – a practice which was not infrequent in dental and radiologic sedation at that time. An addendum to the guideline was produced by the same Committee on Drugs of the AAP 2002 [9] ending the use of the term “conscious sedation” (described above) and clarifying the fact that these guidelines apply to any location where children are sedated – in or out of the hospital. These guidelines use the terminology of “minimal sedation, moderate sedation, deep sedation, and anesthesia.” These levels have been adopted by the ASA, The Joint Commission, and various international organizations. The addendum emphasized that sedatives should only be administered by those skilled in airway management and cardiopulmonary resuscitation [9]. A subsequent iteration of the AAP sedation guidelines was published in December 2006 [10]. For the first time, with the publication of this document, the Joint Commission, ASA, AAP, and the AAPD officially adopted common language to define sedation categories (minimal, moderate, deep, and anesthesia) and the expected physiologic responses for each category. The authors emphasize the idea that sedation is a continuum and that the sedation provider must be capable of rescuing a patient for a level of sedation one step deeper than that which is intended. They recommend “ongoing maintenance of critical skills for airway rescue” and ref-

J. P. Cravero

erence some resources but stop short of specific directions for how best to teach or maintain critical competencies. Deep sedation requires special expertise and personnel resources: Credentials required to administer deep sedation [10]: (i) There must be one person available whose sole responsibility is to constantly observe the patients vital signs, airway patency, and adequacy of ventilation and to either administer drugs or direct their administration. (ii) At least one individual, trained and competent to provide advanced pediatric life support, airway management, and cardiopulmonary resuscitation, must be present. This iteration of the guidelines emphasizes that as the recommendations apply to all sites where sedation is given, clear plans for rescue by emergency medical systems (EMS) must be put in place for settings such as a freestanding clinic or office. The authors included a section on drug interactions and cautions on alternative medications such at St. John’s wort, Kava, and Echinacea and their possible impact on coagulation and sedation provision. This version of the AAP guidelines distinguished monitoring requirements based on the depth of sedation as well as the setting. Pulse oximetry, heart rate, and intermittent blood pressure were recommended during moderate sedation. For deep sedation, “precordial stethoscope or capnography was advised for patients who are difficult to observe (i.e. MRI) to aid in monitoring adequacy of ventilation.” Capnography is “encouraged” but not required, particularly in situations where other means of assessing ventilation are limited. These guidelines maintain the suggestion (from previous versions) that predicting the exact depth of sedation (other than minimal sedation) that will result from the administration of a sedative drug is not possible. In light of this fact, the authors make recommendations on fasting (NPO) durations prior to sedation (Table 2.1) which assume airway protective reflexes could be lost at any time during a moderate or deep sedation and therefore should mimic those that are applied to general anesthesia. Recovery criteria and considerations are also enumerated in these guidelines, including a suggestion for the use of simple “wakefulness” measures as part of the discharge criteria (where a child is simply observed for his/her ability to remain awake for a specified period of time [15–20  min] prior to discharge). The next version of the guidelines AAP Guidelines for Monitoring and Management of Pediatric Patients Before, During, and After Sedation for Diagnostic and Therapeutic Procedures: Update 2016 expanded on the previous versions of these guidelines [2]. The concepts outlined in previous iterations were continued in this version. The authors ­continue to stress the fact that, since the level of sedation may vary over time, providers of sedation must be able to

2  Sedation Policies, Recommendations, and Guidelines Across the Specialties and Continents

rescue patients from a level of sedation that is at least one level deeper than the intended level. So, if the sedation providers intend to deliver deep sedation, they must have the Table 2.1  NPO guidelines

Ingested material Clear liquids: water, fruit juices without pulp, carbonated beverages, clear tea, black coffee Human milk Infant formula Nonhuman milk: because nonhuman milk is similar to solids in gastric emptying time, the amount ingested must be considered when determining an appropriate fasting period Light meal: a light meal typically consists of toast and clear liquids. Meals that include fried or fatty foods or meat may prolong gastric emptying time. Both the amount and type of foods ingested must be considered when determining an appropriate fasting period

Minimum fasting period, h 2 4 6 6

6

Reproduced from American Academy of Pediatrics. American Academy of Pediatric Dentistry et al. [10], with permission from the American Academy of Pediatrics

23

skills required to rescue a patient from general anesthesia. “They must have the ability to recognize the various levels of sedation and have the skills and age and size-appropriate equipment necessary to provide appropriate cardiopulmonary support if needed” [2]. This guideline goes on to be very specific about the competencies required to rescue a child with apnea, laryngospasm, and/or airway obstruction  – including the ability to open the airway, suction, perform bag-mask ventilation, insert an oral airway, nasopharyngeal airway, and laryngeal mask airway, and perform endotracheal intubation. This version of the guidelines is more specific in advising that these competencies are best maintained by frequent simulation and team training for rare events. In addition to these skills, the guidelines advise that (for nonhospital facilities) a protocol for the immediate activation of the EMS system for life-threatening complications must be established and maintained. New to this version of the guidelines is a clear outline of the monitoring and oversight requirements for moderate vs. deep sedation (Table 2.2), which is helpful since the differences are subtle. It is notable that capnography is recom-

Table 2.2  Requirements for moderate versus deep sedation Personnel

Responsible practitioner

Monitoring

Other equipment Documentation

Emergency checklists Rescue cart properly stocked with rescue drugs and age- and siteappropriate equipment (see Appendices 3 and 4) Dedicated recovery area with rescue cart properly stocked with rescue drugs and age- and size-­appropriate equipment (see Appendices 3 and 4) and dedicated recovery personnel; adequate oxygen supply

Moderate sedation An observer who will monitor the patient but who may also assist with interruptible tasks; should be trained in PALS Skilled to rescue a child with apnea, laryngospasm, and/or airway obstruction including the ability to open the airway, suction secretions, provide CPAP, and perform successful bag-valve-mask ventilation; recommended that at least one practitioner should be skilled in obtaining vascular access in children, trained in PALS Pulse oximetry ECG recommended Heart rate Blood pressure Respiration Capnography recommended Suction equipment, adequate oxygen source/ supply Name, route, site, time of administration, and dosage of all drugs administered Continuous oxygen saturation, heart rate, and ventilation (capnography recommended), parameters recorded every 10 min Recommended Required

Deep sedation An independent observer whose only responsibility is to continuously monitor the patient; trained in PALS Skilled to rescue a child with apnea, laryngospasm, and/or airway obstruction, including the ability to open the airway, suction secretions, provide CPAP, perform successful bag-valve-mask ventilation, tracheal intubation, and cardiopulmonary resuscitation; training in PALS is required; at least one practitioner skilled in obtaining vascular access in children immediately available Pulse oximetry ECG required Heart rate Blood pressure Respiration Capnography required Suction equipment, adequate oxygen source/ supply, defibrillator required Name, route, site, time of administration, and dosage of all drugs administered; continuous oxygen saturation, heart rate, and ventilation (capnography required); parameters recorded at least every 5 min Recommended Required

Recommended; initial recording of vital signs may be needed at least every 10 min until the child begins to awaken, then recording intervals may be increased

Recommended; initial recording of vital signs may be needed for at least 5-min intervals until the child begins to awaken, then recording intervals may be increased to 10–15 min

Reproduced from Coté et al. [2], with permission from the American Academy of Pediatrics

24

mended for moderate sedation and required for deep sedation. The guidelines end with a very thorough review of local anesthetics and their pharmacology and recommendations for treatment of local anesthetic toxicity with intralipid. The most recent iteration of the AAP sedation guidelines was published in 2019 [3]. Once again, most of the content is carried over from the previous guidelines – particularly those from 2016. Notably, although the authors recognize that the incidence of aspiration is likely different for sedation activity vs. anesthesia, these guidelines continue the admonition that the NPO intervals for sedation should mimic those of anesthesia since the actual incidence of aspiration is not known for procedural sedation. For emergency procedures, the guidelines advise that the need for the procedure must be weighed against the possible risk imposed by the lack of fasting [3]. In this version of the AAP/AAPD guidelines, there is a recommendation that during deep sedation/general anesthesia for a patient in a dental facility, there must be at least two individuals present with the patient throughout the procedure. Furthermore, the guideline requires both of these individuals to have appropriate training and up-to-date certification in patient rescue. This training should include Pediatric Advanced Life Support (PALS) or Advanced Pediatric Life Support (APLS). The recommendation is that one of the two must be an independent observer who is not performing or assisting with the procedure  – this position can be filled by a physician anesthesiologist, certified registered nurse anesthetist, a second oral surgeon, or a dentist anesthesiologist. Similar recommendations are made for delivery of deep sedation/anesthesia in a hospital or surgicenter setting.

J. P. Cravero

of Dental Anesthesiologists, and the Society of Interventional Radiology. These recommendations are specifically meant to apply to moderate sedation, which is defined (like the AAP guidelines) as a state in which the patient is experiencing a depression of consciousness but is purposefully responsive to voice or light touch, and airway patency should not be impaired. The authors describe a rigorous and systematic review of the literature for these guidelines. Their findings are graded based on the quality of the evidence available to base the recommendations on. Each section of the document also includes a survey of experts in the field intended to augment the conclusions in areas where the published evidence was insufficient to support firm conclusions. These guidelines cover several aspects of sedation – including the pre-­ sedation assessment, monitoring standards (including capnography to supplement standard monitoring of pulse oximetry), personnel requirements (trained observer), a review of medications for sedation, recovery criteria, and required QA/QI processes. While they do not address issues specifically related to children, there is a clear indication, from the inclusion criteria and the searches described, that they are intended to apply to children as well as adults. There is an extensive section on various drug combinations that may be used for moderate sedation. The authors very specifically point out the use of dexmedetomidine and its potential to be substituted for a benzodiazepine when providing moderate sedation. They also take pains to advise careful titration of medications for sedation with the requirement for knowledge of pharmacokinetics in order to avoid stacking of doses and excessive sedation. It is a bit difficult to follow some of the discussion regarding medications since they involve combinations such as propofol and remifentanil, as well as ketamine (and combinations of sedatives with ketamine)  – none of which would seem to apply very well to a set of guidelines addressing only “moderate sedation.” In spite of American Society of Anesthesiologists (ASA) this somewhat incongruous section, the guidelines are clearly written and align with the AAP recommendations in most Policies and Recommendations dimensions. While the ASA has not produced a document specific for The ASA has many other statements that relate to sedapediatric sedation, issues relating to pediatric patients are tion  – they are published on their website at https://www. mentioned or referenced in many of the sedation-related asahq.org/standards-and-guidelines. The statements that perpublications it has produced. Perhaps the most pertinent of tain to sedation include Distinguishing Monitored Anesthesia these documents is the recent “Practice Guidelines for Care from Moderate Sedation Analgesia, Statement on Moderate Procedural Sedation and Analgesia 2018” [11] Granting Privileges for Administration of Moderate Sedation which replaced the previous guidelines titled “Practice to Practitioners, Statement on Granting Privileges to Non-­ Guidelines for Sedation and Analgesia by Non-­anesthesiologist Physicians Supervising Deep Sedation, and Anesthesiologists: An Updated Report by the American the Statement on the Safe Use of Propofol. The ASA also has Society of Anesthesiologists (ASA) Task Force on Sedation produced a number of Expert Consensus Documents; the and Analgesia by Non-Anesthesiologists” [12]. These guide- purpose of these is to disseminate “policies, positions, suglines were developed by a multispecialty group of authors gestions, and definitions to promote the practice of anestheincluding representation from the American Association of siology.” These Expert Consensus Documents are found on Oral and Maxillofacial Surgeons, American College of the same ASA website and include Advisory on Granting Radiology, American Dental Association, American Society Privileges for Deep Sedation to Non Anesthesiologist

2  Sedation Policies, Recommendations, and Guidelines Across the Specialties and Continents

25

Physicians, ASA Physical Classification System, Continuum the education and training requirements for this care along of Depth of Sedation: Definition of General Anesthesia and with the appropriate monitoring and equipment that should Levels of Sedation/Analgesia, and Guidelines for Delineation be present for this type of care. of Clinical Privileges in Anesthesiology. The distinction between sedation, deep sedation, and Several of these statements/guidelines deserve particular monitored anesthesia care (MAC) is frequently misundermention and review. The Statement on Granting Privileges stood. To clarify these definitions, in 2018, the ASA amended for Administration of Moderate Sedation to Practitioners the document entitled Distinguishing Monitored Anesthesia begins by declaring that non-anesthesiologist sedation prac- Care (MAC) From Moderate Sedation/Analgesia (Conscious titioners should supervise moderate sedation only if they are Sedation) to differentiate between the two levels of care. qualified by “education, training, and licensure to administer Important distinctions are noted – that MAC entails an anesmoderate sedation” (It should be noted that some experts thesia assessment and the delivery of sedation by a provider have objected to the term “non-anesthesiologist” since it has who is prepared and qualified to assess and manage physiobeen proposed that this terminology inappropriately includes logical or medical issues as well as to convert to a general physicians of various levels of skill, training, and experience anesthetic. Conversely, patients receiving moderate sedation in one cohort.) [13]. The statement goes on to define various would not be expected to progress to a condition in which the core competencies for the sedation professional including patient could not maintain his/her own airway. obtaining consent, medical history, assessment of risk of In 2019 the ASA published a useful document that aspiration, knowledge of pharmacology of sedative medica- addresses some common (controversial) issues in procedural tions, issues related to oxygen supplementation, proficiency sedation  – under the title Principles for Hospital-based with airway management, knowledge of appropriate moni- Sedation, Analgesia and Anesthesia. This statement is availtors for sedation, knowledge of documentation principles, able at https://www.asahq.org/quality-and-practice-manageand appropriate resuscitation skills. The authors include a ment/quality-improvement/qmda-regulatroy-toolkit/ description of appropriate licensure, practice patterns, and guide-to-anesthesia-department-administration. This docuperformance improvement for these practitioners. ment reviews the US Center for Medicaid and Medicare The Statement on Granting Privileges to Non-­ Services recommendations for sedation oversight. It then Anesthesiologist Physicians for Personally Administering or outlines strategies for Anesthesiology Departments to work Supervising Deep Sedation was updated in October of 2017. with colleagues in other departments to meet the mandates This statement is very brief and worded “Because of the sig- for oversight and quality improvement that are part of the nificant risk that patients who receive deep sedation may standards put forward by the US federal government (CMS). enter a state of general anesthesia, privileges for deep seda- In this rather unique recommendation, the ASA gives a tion should be granted only to non-anesthesiologist physi- point-by-point outline for how anesthesiologists should cians who are qualified and trained in the medical practice of negotiate with colleagues to establish common ground and deep sedation and the recognition of and rescue from general productive discussion on sedation issues. A large portion of anesthesia.” This guideline goes on to advise against non-­ the document specifically addresses a new recommendation anesthesiologists delegating or supervising that administra- from the American College of Emergency Physicians tion of sedation by individuals who are not similarly (ACEP) concerning “unscheduled sedation” (reviewed qualified. At the same time, the ASA Advisory on Granting below) and contrasts the language in that document to the Privileges for Deep Sedation to Non-Anesthesiologist language the ASA has promoted around the same issues. Physicians is a much more detailed document that outlines While there are few firm conclusions for how to resolve the the training, competencies, and licensure that an individual differences between the different organizations’ take on should have in order to deliver deep sedation. This document sedation principles, the presence of a discussion of the variquotes the current CMS statements about practitioners who ous topics involved is evidence of a recognition that are qualified to deliver this form of sedation and the organi- ­conflicting standards exist and that these should be resolved zation for sedation oversight that should be in place in any through dialogue in order to optimize care delivery. organization where this care is being delivered. The ASA “Statement on the Safe Use of Propofol” last amended in 2019 and advises “the involvement of an anes- Center for Medicare and Medicaid Services thesiologist in the care of every patient undergoing anesthe- (CMS) sia is optimal. However, when this is not possible, non-anesthesia personnel who administer propofol should be The US federal government Center for Medicare and qualified to rescue patients whose level of sedation becomes Medicaid Services (CMS) has written The Hospital deeper than initially intended and who enter, if briefly, a state Anesthesia Services Condition of Participation 42 CFR of general anesthesia.” This document goes on to describe 482.52 (a) of 2010 [14] in which it outlines several concepts

26

involved in the delivery of sedation services and how these services should be organized in hospitals that receive reimbursement through CMS. While this is not pertinent to international readers, this agency is critical to reimbursement for US hospitals and organizations; thus its recommendations carry significant weight throughout the country. The guidelines can be found at https://www.cms.gov/Regulations-andGuidance/Guidance/Transmittals/downloads/R74SOMA. pdf. Perhaps the most notable part of this guideline is the recommendation that all anesthesia services be organized under one individual and that the standards for anesthesia care be consistent across an organization. The document goes on to point out that the individual in charge of sedation services would most logically be the Chair of Anesthesiology, but other qualified individuals could fill this position. They outline the various levels of sedation and the distinction between “anesthesia” and “analgesia.” This document also includes very specific language around the need for quality improvement activity with respect to all areas of sedation/ anesthesia and the necessity for a pre-sedation/anesthesia evaluation and a post-sedation/anesthesia evaluation. These CMS guidelines regarding non-anesthesia providers of sedation were revised in January 2011  in the PUB 100–07 “State Operations Provider Certification” which revises Appendix A for various provisions of these recommendations (42 CFR 482.52) concerning anesthesia services. These revisions were made in response to feedback from practitioners. Important changes in these guidelines stem from the CMS acknowledgment that the individual hospitals may establish their own policies and procedures with respect to the qualifications of analgesia providers and the clinical situations which distinguish anesthesia from analgesia. The policies must follow nationally recognized guidelines and can include guidelines of one or more specialty societies.

The Joint Commission Issues relating to sedation regulations and guidelines (in general) and pediatric sedation (in specific) are found in a variety of locations in the The Joint Commission Handbook and website (http://www.jointcommission.org). The JCAHO 2004 Comprehensive Accreditation Manual for Hospitals was intended to set the standards for sedation and anesthesia care for patients in any setting. The Joint Commission recommendations are important when considering the credentialing and privileging of sedation providers. They require hospitals define the scope of practice for practitioners. It is important to distinguish the term “credentialing” from “privileging.” “Credentialing” is the process whereby designated hospital appointees assure that physicians who work in the hospital have the appropriate education, training, and licensure to practice in the institu-

J. P. Cravero

tion. “Privileging” specifically gives permission to hospital staff to provide care in various clinical settings or perform particular procedures in a given institution. With regard to sedation privileging, each healthcare facility is mandated by The Joint Commission to approve a plan to provide sedation and anesthesia care. Each institution must outline the criteria for determining which practitioners are qualified to provide the service. It is important to recognize the evolution of the role of the Anesthesiology Department in the delivery of sedation as outlined by The Joint Commission. Earlier Joint Commission publications placed responsibility for sedation oversight directly on the Department of Anesthesiology and its chairman. Subsequent revisions of this document have revised the language: the Anesthesiology Department play an important advisory role but is not directly responsible for sedation care, privileging, or quality assurance. In the current Joint Commission manual, there are recommendations for the training that may be provided for other sedation providers: “Individuals administering moderate or deep sedation and anesthesia are qualified and have the appropriate credentials to manage patients at whatever level of sedation or anesthesia is achieved, either intentionally or unintentionally.” Referring specifically to deep sedation it states, “individuals must be qualified to rescue patients from general anesthesia and are competent to manage an unstable cardiovascular system as well as a compromised airway and inadequate oxygenation and ventilation.” It goes on “Each organization is free to define how it will determine that the individuals are able to perform the required types of rescue. Acceptable examples include, but are not limited to, ACLS certification, a satisfactory score on a written examination developed in concert with the department of anesthesiology, a mock rescue exercise evaluated by an anesthesiologist.” Although the Joint Commission still believes that Anesthesiology Departments should play a role in the development of training and privileging programs for sedation, they no longer hold the central role of being “in charge” of sedation services. Key roles in sedation oversight may be filled by qualified specialists of many different subspecialties.

 merican College of Emergency Physicians A (ACEP) Guidelines The American College of Emergency Medicine (ACEP) has put forward a wide range of statements, clinical practice advisories, and clinical policy statements concerning sedation of children for procedures. The 2019 American College of Emergency Physicians Policy Compendium includes a statement Procedural Sedation in the Emergency Department which has been updated several times since it was first pub-

2  Sedation Policies, Recommendations, and Guidelines Across the Specialties and Continents

lished in 1992 (https://www.acep.org/globalassets/new-pdfs/ policy-statements/procedural.sedation.in.the.ed.pdf). This statement emphasizes the importance of procedural sedation – improving the quality and safety of emergency department care. It mentions a variety of medications and non-pharmaceutical interventions that improve care during procedures. Importantly, this document clearly states that “NPO status has not been demonstrated to reduce risk of emesis or aspiration in ED procedural sedation.” It also establishes that “the American College of Emergency Physicians is the authoritative body for the establishment of guidelines for sedation in emergency department patients.” This could be interpreted as a challenge to the CMS guidelines (outlined above) which places oversight for all sedation at a given institution under one individual. There are multiple other sedation statements from this organization. In 2005 and 2014, the ACEP published Clinical Policy: Procedural Sedation and Analgesia in the Emergency Department [4, 15]. Similar to the ASA guidelines, the ACEP guidelines apply to all patients, (adults and children) who receive sedation. They were developed based on a structured literature review, and the recommendations are graded based on the strength of the evidence. They recognize that sedation is a continuum and maintain that practitioners should possess competence in cardiovascular resuscitation and airway management which should include a patient who has achieved general anesthesia. ACEP considers these skills, including the administration of propofol and deep sedation, to be a fundamental part of the emergency medicine training curriculum and inclusive of the training required of all board-­ certified emergency physicians [15, 16]. The ACEP guidelines differ slightly from those of the AAP and ASA with respect to NPO guidelines. Both the AAP and ASA recommend fasting intervals for elective cases similar to those required for general. Those guidelines do not make recommendations specifically for the nonelective sedation case. As mentioned above, emergency medicine sedation providers must cope with patients who do not meet appropriate NPO criteria and are not having “elective” procedures. In the last 20 years, there have been several studies in the emergency medicine literature that have reported very low rates of aspiration or pulmonary complications in patients who were sedated without meeting the NPO recommendations from the AAP or ASA [17–19]. Previous publications from the ACEP have concluded that there is insufficient evidence to conclude that fasting actually changes outcome for sedation (see above) [20]. This clinical policy recommends that providers “do not delay procedural sedation in adults or pediatrics in the ED based on fasting time.” It also includes a recommendation in favor of capnography for procedural sedation and recommends the use of various potent sedatives in emergency department practice [4].

27

In 2007 ACEP produced a guideline specifically addressing the issue of fasting prior to sedation [21]. This clinical practice advisory is titled “Fasting and Emergency Department Procedural Sedation and Analgesia: A Concensus-Based Clinical Practice Advisory.” The paper begins with an extensive review of the guidelines that have been set forth by the ACEP, AAP, and ASA concerning NPO status and considers them in the context of the emergency department setting. This consensus-based clinical advisory concludes that there is actually scarce literature to document the perceived risk that various NPO times pose with respect to sedation complications. The authors suggest that the issue of NPO interval needs to be considered in the context of the urgency and duration of the procedure as well as the risk stratification of the patient, nature of food intake, and depth/ type of sedation targeted. The result is a somewhat complex strategy that weighs NPO time vs. emergent/urgent/semiurgent nature of the case vs. duration of the procedure. Figure  2.1 schematically describes the recommendations that result from these guidelines [21]. These NPO recommendations conclude that “recent food intake is not a contraindication for administering procedural sedation and analgesia, but should be considered in choosing the timing and target level of sedation” [15, 21]. In 2004 ACEP published evidence-based guidelines on the use of specific medications for use in pediatric sedation. Clinical Policy: Evidence-Based Approach to Pharmacologic Agents Used in Pediatric Sedation and Analgesia in the Emergency Department [22] was a thorough document that has not been updated and (perhaps) replaced by separate clinical policy statements on the use of drugs such as propofol and ketamine (see below). Another well-researched publication, Clinical Policy: Critical Issues in the Sedation of Pediatric Patients in the Emergency Department [20] was published 4 years later. The “critical issues” statement supported earlier recommendations on NPO status and reviewed the use of sedatives such as nitrous oxide, chloral hydrate, and sucrose. Their recommendations have been accepted by a wide range of surgical and nursing organizations and have been published in corresponding journals [23, 24]. The ACEP website includes a position statement titled Procedural Sedation and Analgesia in the Emergency Department: Recommendations for Physician Credentialing, Privileging, and Practice. https://www.acep.org/globalassets/uploads/uploaded-files/acep/clinical-and-practice-management/resources/sedation/acep-sedation-position.pdf. This document defines various states of sedation/anesthesia and goes on to state that “Graduates of emergency medicine residency and fellowship programs…are qualified for all forms of analgesia and all levels of sedation in all ages.” For providers who are not trained in emergency medicine, it advises that the chief of the unit (or section) will need to set

28

J. P. Cravero

Standard-risk patienta Oral intake in the prior 3 Emergent hours Procedure All levels of Nothing sedation

Procedural Urgencyb Urgent SemiProcedure Urgent All levels of sedation

All levels of sedation

All levels of sedation

Procedural sedation and analgesia targeted depth and durationc Minimal sedation only

NonUrgent

Clear liquids only

All levels of sedation

All levels of sedation

Up to and including brief deep sedation

Up to and including extended moderate sedation

Light snack

All levels of sedation

Up to and including brief deep sedation

Up to and including dissociative sedation; non-extended moderate sedation

Minimal sedation only

Heavier snack or meal

All levels of sedation

Up to and including extended moderate sedation

Minimal sedation only

Minimal sedation only

Higher-risk patienta Oral intake in the prior 3 Emergent hours Procedure All levels of Nothing sedation

Dissociative sedation; brief or interm ediate-length moderate sedation Extended moderate sedation

Procedural Urgencyb Urgent Procedure

Semi-Urgent

Non-Urgent

All levels of sedation

All levels of sedation

All levels of sedation

Clear liquids only

All levels of sedation

Up to and including brief deep sedation

Up to and including extended moderate sedation

Minimal sedation only

Light snack

All levels of sedation

Up to and including dissociative sedation; non-extended moderate sedation

Minimal sedation only

Minimal sedation only

Heavier snack or meal

All levels of sedation

Up to and including dissociative sedation; non-extended moderate sedation

Minimal sedation only

Minimal sedation only

Brief deep sedation

Intermediate or extended-length deep sedation Brief: 20 minutes

Fig. 2.1  ACEP NPO considerations and aspiration risk. (Reproduced from Green et al. [21]. Epub 2006 Nov 1. Reprinted with permission from Elsevier)

the standards for training and proctoring of practice prior to privileging these practitioners for sedation practice. ACEP recently updated a clinical practice advisory on propofol use in the emergency department [16, 25]. This guideline is consistent with other ACEP publications in that it does not consider lack of an NPO interval as a contraindication to administration of propofol. It notes providers who deliver propofol sedation must be qualified for deep sedation. As a departure from the AAP or ASA guidelines, this practice advisory allows for a single provider to both deliver sedation with propofol and perform the procedure – as long as that individual is “prepared to interrupt the procedure to perform resuscitation.” There are also some recommendations for dosing of propofol as a single agent and in combination with ketamine or opioids. ACEP also has a practice guideline on ketamine use in the emergency department which was originally written in 2004 and updated in 2011 [26, 27]. The authors point out the unique sedative qualities of ketamine and have a separate “level” of sedation termed “dissociative” sedation as applied to the state that is induced by ketamine. (This definition is not recognized by other major organizations that have issued

guidelines on sedation practice). Because of its unique properties, the authors argue a separate clinical policy is needed outside of other recommendations on sedative administration in the emergency department [22]. This document outlines best use (minor painful procedures) and enumerates some possible contraindications (“airway instability”). Two providers are recommended for dissociative sedation, although IV access is unnecessary when the drug is given by the intramuscular route. Recommendations for recovery and discharge are also included. In 2019, ACEP published a guideline specifically aimed at addressing issues related to the administration of ­unscheduled procedural sedation. Titled Unscheduled Procedural Sedation: A Consensus Practice Guideline, this extensive document reviews most issues associated with procedural sedation with special attention to the unscheduled case situation. The guideline was based on clinical analysis of existing literature between 2000 and 2018 and endorsed by multiple organizations (Fig.  2.2) [28]. Most of the document’s 21 pages outline concepts that are similar to the ACEP recommendations for general procedural sedation. Two sedation providers are required, a sedation provider and a sedation

2  Sedation Policies, Recommendations, and Guidelines Across the Specialties and Continents Fig. 2.2  Organizations that approved the ACEP consensus practice guideline on unscheduled sedation. (Reproduced from Green et al. [28] with permission from Elsevier)

29

Organizations that participated and endorsed the guideline • American College of Emergency Physicians • American Academy of Emergency Medicine • American Board of Emergency Medicine • American College of Cardiology • American College of Medical Toxicology • American College of Osteopathic Emergency Physisians • Association of Academic Chairs of Emergency Medicine • Emergency Medicine Residents’ Association • Emergency Nurses Association • Society for Academic Emergency Medicine • Society for Pediatric Sedation Organizations that participated and provided input • American Academy of Pediatrics • American Academy of Pediatrics Section on Critical Care • American Academy of Pediatrics Section on Pediatric Emergency Medicine • American Society for Gastrointestinal Endoscopy • Council of Emergency Medicine Residency Directors • American Association of Oral and Maxillofacial Surgeons • Society of Critical Care Medicine • Society of Interventional Radiology Organizations that participated review comments • American Association of Nurse Anesthetists Eigth other organizations representing general medicine, anesthesiology, dentistry, and gastroenterology were invited to participate, but either declined or did not respond.

monitor. The recommendations in this document vary slightly from the AAP, and ASA guidelines in that the qualifications for the “procedural sedation monitor” are less complex and not directed at the specific level of sedation. This individual is described as “a nurse, respiratory therapist, or other health care professional who is privileged based upon local oversight, training, and verification of skills.” These skills required are limited to monitoring the patient, being able to assist the sedation provider (who may be performing the procedure) in resuscitation and summon to effectively summon help. Furthermore they allow for some dual tasking of the sedation monitor – “the sedation monitor can assist with minor, interruptible tasks as long as they do not materially interfere with the effective procedural sedation monitoring.” Given the nature of the unscheduled procedures addressed in this document and the lack of data relating aspiration to NPO duration, the authors conclude that given the exceptionally low risk of pulmonary aspiration with procedural sedation and absent evidence of an impact from fasting, reform is appropriate for recommendations regarding pre-procedural oral intake [28].

I nternational Committee for the Advancement of Procedural Sedation Consensus Statement on Fasting for Procedural Sedation Almost all of the general guidelines on sedation that have been published include some mention of the duration of fasting that is required prior to providing procedural sedation. As outlined above in the AAP, ASA, and ACEP section of

this chapter, these recommendations do not always align perfectly. This is largely due to the lack of very high-quality data on the topic. To attempt to address this issue and other topical issues in sedation, an International Collaborative of experts were formed, the International Committee for the Advancement of Procedural Sedation (ICAPS). This group used a review of the available literature, along with a Delphi methodology, to come up with a set of consensus recommendations regarding fasting duration prior to sedation [29]. These experts concluded that there is “no association between aspiration and compliance with fasting guidelines.” They suggest that fasting guidelines for procedural sedation should be less restrictive than those for general anesthesia, and their suggested algorithm for fasting prior to procedures is presented in Fig. 2.3.

 merican Dental Association Sedation A Guidelines As mentioned in the section on the AAP guidelines on sedation (above), the American Academy of Pediatric Dentistry (AAPD) has been involved in the writing and dissemination of the AAP/AAPD [3] guidelines on sedation from their inception. Most of the versions of the AAP guidelines have been co-written with the AAPD over the years – including the current version. The most important addition in the current iteration is the inclusion of the recommendation that two providers with specific sedation rescue training and credentials (Pediatric Advanced Life Support) should be present for

30

J. P. Cravero Pre-sedation assessment – risk factors Negligible risk factors

Moderate risk factors

Mild risk factors Patient • Severe systemic disease • Moderate obesity1 • Age 12 months or less • Hiatal hernia

• No risk factors shown to the right

Patient • Severe systemic disease that is a constant threat to life • Severe obesity1, obstructive sleep apnoea • Airway abnormalities2 • Hyperemesis, oesophageal disorders3, bowel obstruction4

Procedure/Sedation • Upper endoscopy • Bronchoscopy • Propofol principal sedative

Procedure/Sedation • Anticipated need for assisted ventilation or other advanced airway management

Elective procedures Negligible aspiration risk

Clear liquids5 Breast milk Food, formula, nonhuman milk

Fasting approximately 2 h6

Unrestricted

Unrestricted

h6

Fasting approximately 4 h6

Fasting approximately 4 h6

Fasting approximately 6 h6

Fasting approximately 2

Unrestricted Fasting approximately 2

Moderate aspiration risk (any of the above) Refer for anaesthesia care

Mild aspiration risk (any of the above)

h6

Urgent or emergency procedures

No delay based on fasting time

No delay based on fasting time

No delay based on fasting time. Anaesthesia care if available; if not consider ketamine as sole sedative agent.

Fig. 2.3  Pre-procedural sedation algorithm from the International Committee for the Advancement of Procedural Sedation. (Reproduced from Green et al. [29] with permission from John Wiley and Sons)

sedation in a dental office. This is a significant change from the earlier versions which only recommended one such qualified sedation provider to be present for sedation of this kind. In addition to the collaborative guidelines (above), the American Dental Association (ADA), independently, published general guidelines regarding sedation titled “Guidelines for the Use of Sedation and General Anesthesia by Dentists.” They were last updated in October of 2016, and they are available at https:// www.ada.org/~/media/ADA/Education%20and%20Careers/ Files/ADA_Sedation_Use_Guidelines.pdf. The guideline outlines depths of sedation consistent with that described by the AAP/AAPD and the ASA. It contains descriptions of routes of administration for sedative medications, ASA classification for sedation patients, and monitoring guidelines for sedated patients. There is a very specific outline of the educational requirements for dentists regarding various levels of sedation, including specific programs and life support training. In this regard the guidelines are more detailed than those provided by other organizations. Deep sedation requires the presence of a minimum of three individuals: one dentist who is credentialed to administer deep sedation or anesthesia and two additional personnel who have current certification in Basic Life Support (BLS) Course for the

Healthcare Provider. There are two pathways for dentists to qualify for deep sedation certification: (1) completion of an advanced education program on the administration and management of deep sedation or anesthesia, which must be accredited by the ADA Commission on Dental Accreditation and a current certification in both BLS for Healthcare Providers and Advanced Cardiac Life Support (ACLS) or (2) an appropriate dental sedation/anesthesia emergency management course. This guideline goes on to recommend that dentists administering deep sedation or general anesthesia must remain within the facility until the patient meets discharge criteria (or is discharged) and must monitor the patient continuously until the patient meets the criteria for recovery. Those who provide pediatric sedation must have Pediatric Advanced Life Support (PALS) in addition to directed pediatric training and education. This set of ADA guidelines is presented in sections, divided by sedation level: minimal, moderate, and deep sedation sections. Specific recommendations are given for training of sedation providers, preoperative preparation of patients, monitoring and documentation, recover and discharge criteria, and personnel/equipment requirements. The document is intended for adults and for children 12 years of

2  Sedation Policies, Recommendations, and Guidelines Across the Specialties and Continents

age and over. The ADA refers to the (AAP/AAPD) Guidelines for Monitoring and Management of Pediatric Patients During and After Sedation for Diagnostic and Therapeutic Procedures for guidelines concerning sedation of young children, infants, and toddlers [30]. These guidelines address some issues unique to the office-based dental practice and to the special needs child. If the dental patient undergoing deep sedation or general anesthesia is mentally and/or physically challenged, it may not be possible to have a comprehensive physical examination or appropriate laboratory tests prior to administering care. In these situations, the dentist responsible for administering the deep sedation or general anesthesia should document the reasons preventing the recommended preoperative assessment prior to administering sedation [10]. Nitrous oxide is a recognized and acceptable sedative, alone or in combination with other sedatives.

Table,

31

In 2018 AAPD published an update of its 2012 Guideline on Use of Anesthesia Personnel in the Administration of Office-based Deep Sedation/General Anesthesia to the Pediatric Dental Patient [31]. It affirms the fact that there are several categories of pediatric patients, such as those with developmental delays and autism, who require deep sedation for dental interventions. Further, this manuscript recognizes that when sedation/anesthesia care is provided in the dental office, it is much more cost-effective and convenient to schedule than when it is delivered in a large hospital setting. This guideline is careful to define the aspects of training that are required in order to deliver this care. Specifically, this policy details the different types of sedation and anesthesia providers along with their permissible responsibilities and training (Fig. 2.4). Emergency preparedness must be updated and practiced on a regular basis, and recovery must be moni-

ANESTHESIA EDUCATION AND TRAININGCOMPARISON

Anethesia provider

Permitted to function independent of supervivion by anesthesiologist

Mininum duration of program required for certification

Min. # of DAGA cases

Min. # of pediatric cases

Definition of pediatric patient

Min. # of DAGA cases involving patients with SHCN

No

24 months

400GA cases

50

0-18

N/A

National Commission for Certification of Anesthesiologist Assistants

In some states

24 months

25/400

 verbal ability 4–10 Development of reasoning Elimination of egocentrism Improved verbal communication 10+

Can think abstractly Future thinking Heightened self-consciousness

Strategy for preprocedure preparation Use real objects to help child master the situation Reinforce good behavior Keep parent with child as much as possible Allow time for questioning Provide detail Use concrete teaching materials and simple medical terms Involve patient in decision-making Provide information in advance Support need for self-control and independence Offer explanations in clear, technical terms Respect privacy and self-image concerns

Adapted from [20]

example, stating “this may feel like a pinch” or “some children say this feels warm and tingly” gives children a sensory as opposed to negative focus. Positive reinforcement such as “you are being brave” or “you did a good job of holding still” is a nice way of providing encouragement or praise. Finally, children are often very concrete thinkers. Stating that “the nurse is going to draw your blood” is too vague for most children to understand. Rather, describing the procedure in a stepwise fashion (e.g., “the nurse is going to clean your arm, you will feel a cold pad to wash your skin, we will use this tourniquet to give your arm a hug, etc.”) provides both sensory and detailed information that allow the children a greater sense of control [18].

Procedures A pediatrician will encounter many different common procedures that may require procedural sedation. Depending on the procedure, a patient may require analgesia or sedation/anxiolysis or both. For instance, an infant who needs a head MRI will likely require a sedative agent, while a cooperative adolescent may only require pain medication for a lumbar puncture. On the other hand, a child with an angulated forearm fracture will need both analgesia and sedation for the reduction of the fracture. It is difficult to characterize procedures to predict the medication require-

45 Table 3.4  Procedures that may require procedural sedation Radiologic imaging procedures (e.g., CT scan, MRI, ultrasound) Laceration repair Lumbar puncture Foreign body removal Abscess management (e.g., incision, drainage, and packing) Burn or wound debridement Relocation of a dislocated joint Fracture reduction Joint aspiration Prepubescent gynecologic examination Hernia reduction Peripherally inserted central catheter (PICC) placement Bone marrow aspiration Central line placement Thoracentesis Chest tube placement Cardioversion Endotracheal intubation CT computed tomography, MRI magnetic resonance imaging

ment. The temperament, cognitive development, and patient’s past experience will alter what is needed for any given procedure. Table 3.4 lists the most commonly encountered procedures that may require procedural sedation. This list is not intended to be inclusive nor exhaustive. For instance, some very common procedures may require procedural sedation in a minority of patients (e.g., venipuncture). Additionally, there are some procedures on the list (e.g., endotracheal intubation, thoracentesis) that most general pediatricians will not perform once they have completed residency training. While the choice of agents is covered in great detail in other chapters, there are a few points that bear repeating. It should be noted that while opioids do have some sedative effects, sedation often enhances analgesic efficacy. In a patient who is anxious or stressed, concomitant treatment with a sedative may reduce the needed dose of narcotics. Furthermore, the use of local and regional anesthetics (e.g., nerve blocks) may reduce the total dose of sedatives and analgesics required.

Other Considerations Given the large number of resources required to safely perform procedural sedation, only primary care pediatricians in a hospital or medical center setting will likely be able to perform procedural sedation for their patients. However, this does not mean that pediatricians outside of these settings cannot assist and advise in the sedation of their patients. Our understanding of these patients and the process will allow us to play an integral role in the planning and implementation of the sedation.

46

As previously stated, it would be extremely unlikely that procedural sedation becomes common for painful procedures such as phlebotomy or IV placement. Local anesthetics can dramatically lessen the pain associated with procedures that require penetration of the skin [23]. In general, there are three processes by which the local anesthetic is delivered to the skin. The anesthetic can be injected locally via a small-gauge needle; it can diffuse passively through the skin via a cream or gel or be administered by a needleless system that enhances passage of the local anesthetic through the skin (e.g., heat-enhanced diffusion, iontophoresis, sonophoresis, laser-assisted passage, or pressurized gas delivery) [24]. Another topical treatment to reduce pain is the use of a vapocoolant spray. By rapidly cooling the skin, it is thought that initiation and conduction of nerve impulses are reduced and the refractoriness is increased [24]. A differentiating feature of these different methodologies is the timing and onset of anesthesia. Finally, less invasive routes, such as intranasal administration, allow for the delivery of both analgesics and anxiolytics without the need for intravenous access [25, 26]. There have been a number of studies that have demonstrated the effectiveness of distraction as a technique to minimize pain and anxiety around painful medical procedures [27] (Refer to Chaps. 40 and 41). While there are several postulated theories as to how distraction works to reduce pain, there is much anecdotal evidence to suggest that it is an excellent pain-management intervention. Child life therapists are another excellent resource to assist in pain management, both in regard to preparing for a procedure and providing distraction during a procedure [26]. Even proper positioning can assist in making a painful procedure less traumatic [18]. Depending on the procedure, sitting on a parent’s lap or allowing a child to hold a parent’s hand can help reduce procedure-related anxiety. For young infants, skin-to-­ skin contact, non-nutritive sucking, and sucrose water have been demonstrated to be helpful in reducing perceived pain and should be considered for certain procedures when medically allowable.

Future Directions One of the most recent advances in our understanding of adverse reactions due to medication use is due to our increasing knowledge of pharmacogenetics. The observed differences in response between patients to the same dose of the same drug likely are attributed to how a given individual metabolizes a given agent. For instance, differences in the level of cytochrome P-450-dependent monooxygenase activity may result in differences in both efficacy and toxicity of certain agents [28]. As an example, variants in the genotype CYP2D6 likely explain different responses to codeine, including potentially life-threatening toxicity as

V. W. Chiang and M. S. Siddiqui

the result of accumulation of its active metabolites [28]. In the future our understanding of pharmacogenetics will likely be integrated into the decision-making process as we choose agents to provide procedural sedation in the safest manner possible.

Conclusion In summary, it is the responsibility of the sedation provider to advocate for his/her own patients, especially with regard to pain, fear, and anxiety that may accompany a medical procedure. Being an advocate does not mean that all patients should be sedated for every painful procedure. In reality, the provider must balance the pain associated with the procedure with what is safest for the patient. In essence, the approach to procedural sedation is as much about choosing when not to sedate as it is to tailoring the sedation to the patient and procedure.

Case Studies Case 1 You are in your office when a father brings his son in for evaluation. He is a healthy 14 year old with no significant past medical history who accidentally slammed his finger in a door. You obtain X-rays, which are negative. However, he has a large subungual hematoma that is moderately painful to touch. What would be your approach to managing pain for the treatment of the hematoma? Considerations  The treatment for a subungual hematoma involves draining the hematoma by placing a hole in the nail itself (trephination). Since the nail is not innervated, this is a relatively painless procedure and, in general, does not require any analgesia. However, there are still patients who may be quite anxious. Most 14 year olds can be reasoned with, for example, pointing out that it does not hurt when one clips one’s nails. One can also position one’s body between the patient and the affected finger during the procedure so as to “prevent” the patient from seeing the procedure itself. In extreme cases, the use of a short-acting anxiolytic may be warranted. Case 2 You and a nurse are together seeing urgent patients for your clinic. A mother brings her 2-year-old son in for blood work. She is here because she herself is quite needle phobic and thinks that her son is as well. She would like her son to be sedated for the blood draw, and the local lab said that they do not sedate patients for phlebotomy. What would your approach be to this patient?

3  Procedural Sedation: Let’s Review the Basics

Considerations  Whenever a patient undergoes procedural sedation, one has to weigh the risks and benefits. In general, phlebotomy is not considered a typical procedure for which procedural sedation is used. That being said, it does not mean one should not try to minimize the discomfort associated with the procedure. For 2 year olds, depending on the urgency, one could consider the use of a topical anesthetic prior to a blood draw. Also, this is a great age where distraction techniques may help as well. Case 3 Your office is in a small medical center that shares a procedure room where you can provide procedural sedation. The procedure room is well stocked, including having a pediatric code cart that is kept up to date. You and your nurses have done a number of procedures there, and in general you feel quite comfortable providing procedural sedation. One of your 2-year-old patients is brought in after a fall through a glass coffee table. The patient has multiple deep lacerations to both forearms, which will require significant repair. Of note, the patient has trisomy 21. What would your approach be to this patient? Considerations  The provision of procedural sedation is not simply about providing the medications, but also managing the potential complications that may occur. While this patient may tolerate sedation without any difficulty, there are other factors to consider. Patients with trisomy 21 often have macroglossia and hypotonia, which can increase the difficulty of managing the patient’s airway should hypoventilation or apnea occur. Additionally, patients with trisomy 21 can also have complex congenital heart disease, which can affect which agents are chosen for the procedural sedation. At the very least, consultation with an expert in procedural sedation, if not transfer to a facility with even more resources, should be considered.

References 1. American Academy of Pediatrics Committee on Drugs. Section on anesthesiology: guidelines for the elective use of conscious sedation, deep sedation, and general anesthesia in pediatric patients. Pediatrics. 1985;76:317–21. 2. Walco GA. Needle pain in children: contextual factors. Pediatrics. 2008;122(Suppl 3):S125–9. 3. Kennedy RM, Luhmann J, Zempsky WT.  Clinical implications of unmanaged needle-insertion pain and distress in children. Pediatrics. 2008;122(Suppl 3):S130–3. 4. Kaplan RF, Cravero JP, Yaster M, Coté CJ. Sedation for diagnostic and therapeutic procedures outside the operating room. In: A practice of anesthesia for infants and children: Elsevier Inc; 2009. p. 1023–48. https://doi.org/10.1016/B978-141603134-5.50052-4. 5. Krauss B, Green SM.  Procedural sedation and analgesia in children. Lancet. 2006;4;367(9512):766–80.

47 6. Coté CJ, Wilson S, American Academy of Pediatrics, American Academy of Pediatric Dentistry. Guidelines for monitoring and management of pediatric patients before, during, and after sedation for diagnostic and therapeutic procedures. Pediatrics. 2019;143(6):e20191000. 7. Pacheco GS, Ferayorni A. Pediatric procedural sedation and analgesia. Emerg Med Clin North Am. 2013;31(3):831–52. https://doi. org/10.1016/j.emc.2013.04.002. Epub 29 June 2013. 8. Coté CJ. “Conscious sedation”: time for this oxymoron to go away! J Pediatr. 2001;139(1):15. 9. American Society of Anesthesiologists Task Force on Sedation and Analgesia by Non-Anesthesiologists. Practice guidelines for sedation and analgesia by non-anesthesiologists. Anesthesiology. 2002;96:1004. 10. Krauss B, Green SM. Sedation and analgesia for procedures in children. N Engl J Med. 2000;342:938–45. 11. Cote CJ, Wilson S.  The work group on sedation. Guidelines for monitoring and management of pediatric patients during and after sedation for diagnostic and therapeutic procedures: an update. Pediatrics. 2006;118:2587–602. 12. Cravero JP, Blike GT, Beach M, et  al. Incidence and nature of adverse events during pediatric sedation/anesthesia for procedures outside the operating room: report from the pediatric sedation research consortium. Pediatrics. 2006;118:1087–96. 13. Cote CJ, Karl HW, Notterman DA, et al. Adverse sedation events in pediatrics: analysis of medications used for sedation. Pediatrics. 2000;106:633–44. 14. Cote CJ, Notterman DA, Karl HW, et al. Adverse sedation events in pediatrics: a critical incident analysis of contributing factors. Pediatrics. 2000;105:805–14. 15. Flood RG, Krauss B.  Procedural sedation and analgesia for children in the emergency department. Emerg Med Clin North Am. 2003;21:121–39. 16. Joint Commission on Accreditation of Healthcare Organizations. Comprehensive accreditation manual for hospitals: the official handbook by the JCAHO. Oakbrook Terrace: Joint Commission on Accreditation of Healthcare Organizations; 2005. 17. Henderson K, Womack W. Noninvasive monitoring for procedural sedation. In: Krauss B, Brustowicz RM, editors. Pediatric procedural sedation and analgesia. Philadelphia: Lippincott, Williams & Wilkins; 1999. 18. Standards for Basic Anesthetic Monitoring. Committee of Origin: standards and practice parameters (Approved by the ASA House of Delegates on October 21, 1986, and last amended on October 20, 2010 with an effective date of July 1, 2011). http://www. asahq.org/ForHealthcare-Professionals/Standards-Guidelines-andStatements.aspx. 19. Cohen LL. Behavioral approached to anxiety and pain management for pediatric venous access. Pediatrics. 2008;122(Suppl 3):S134–9. 20. Jaanist T, Hayes B, von Bayer CL. Providing children with information about forthcoming medical procedure: a review and synthesis. Clin Psychol Sci Pract. 2007;14:124–43. 21. Moynihan R, Kurker C.  The perioperative environment and the pediatric patient. In: Ferrari LR, editor. Anesthesia and pain management for the pediatrician. Baltimore: The Johns Hopkins University Press; 1999. 23. Zempsky WT.  Pharmacologic approaches for reducing venous access pain in children. Pediatrics. 2008;122(Suppl 3):S140–53. 24. Burke D, Mogyoros I, Vagg R, Kiernan MC. Temperature dependence of excitability indices of human cutaneous afferents. Muscle Nerve. 1999;22:51–60. 25. Grassin-Delyle S, Buenestado A, Naline E, Faisy C, Blouquit-Laye S, Couderc LJ, Le Guen M, Fischler M, Devillier P. Intranasal drug delivery: an efficient and non-invasive route for systemic administration: focus on opioids. Pharmacol Ther. 2012;134(3):366–79.

48 26. Wolfe TR, Braude DA.  Intranasal medication delivery for children: a brief review and update. Pediatrics. 2010;126(3): 532–7. 27. Leahy S, Kennedy RM, Hesselgrave J, et  al. On the front lines: lessons learned in implementing multidisciplinary peripheral

V. W. Chiang and M. S. Siddiqui venous access pain-management programs in pediatric hospitals. Pediatrics. 2008;122(Suppl 3):S161–70. 28. Gasche Y, Daali Y, Fathi M, Chiappe A, Cootini S, Dayer P, et al. Codeine intoxication associated with ultrarapid CYP2D6 metabolism. N Engl J Med. 2004;351:2827–31.

4

Pre-sedation Assessment Timothy Horeczko and Mohamed Mahmoud

Introduction In this chapter we will present the essentials of pre-sedation screening and risk stratification, discuss fasting guidelines, and review the most commonly encountered scenarios and comorbidities that impact sedation management and outcomes. Today’s practice of pediatric sedation (PS) involves ever more complex patients whose care is coordinated with multidisciplinary teams. Technological advances have allowed for the development of various invasive and noninvasive pediatric procedures and imaging modalities, resulting in a tremendous demand for and growth in PS in children. Despite the increasing complexity and patient volume, sedation providers generally meet the child and his family only minutes before the scheduled (or unscheduled) procedure. The provider must assess the situation quickly and accurately to ensure safety and optimal effectiveness. Important data from all available resources should be gathered and synthesized before the procedure to formulate a successful sedation plan within the context of the urgency of the procedure. Sedation providers must work to not only to optimize patients prior to the procedure but should also act as gatekeepers for procedural safety. The saying “an ounce of prevention is worth a pound of cure” encapsulates the pre-sedation mindset. The main objective for the sedation provider during pre-procedural assessment is to answer the question: is this child optimized for the procedure or not? Components of a successful sedation plan include readily accessible medical records; a thorough medical history with review of systems and careful attention to red flags, pre-­ T. Horeczko (*) Department of Emergency Medicine, Harbor—UCLA Medical Center, Torrance, CA, USA e-mail: [email protected] M. Mahmoud Department of Anesthesia, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA e-mail: [email protected]

sedation tests, or consultation if indicated; a targeted physical exam; and a complete understanding of the procedure and its potential physiologic effects on the patient.

Pre-sedation Screening All children scheduled for elective sedation should receive a pre-screen telephone call before the scheduled invasive or noninvasive procedure. Last-minute cancellation due to new information surfacing on the day of the procedure can result in delay of care and economic loss for the parents and the institution. The telephone screening allows for review of the medical history, gives the opportunity to determine if there is some underlying medical issue that requires further investigation, confirms that the child has not been recently ill, and reinforces nil per os (NPO) instructions. Pertinent data points should be clearly documented and attached to a standardized, hospitalapproved sedation assessment form. Once the screening process is complete, an established triage system can help to determine whether the procedure is appropriate for non-anesthesiologist sedation or whether the expertise of an anesthesiologist is needed. In many centers there is a “point person” to whom non-anesthesiologists may direct questions regarding patient management issues in off-site venues. This coordinator should be familiar with the requirements, challenges, and needs of the individual specialists. In the case of an urgent or emergent (nonelective) procedure, the same logic applies: gather as much information as possible and reasonable for your setting to make the most informed decision regarding the timing and approach to the procedure.

History The process of constructing a successful sedation plan starts with a careful, targeted history focusing on a few critical domains. Ask about past problems or known abnormalities of the respiratory, cardiovascular, neurologic, gastrointesti-

© Springer Nature Switzerland AG 2021 K. P. Mason (ed.), Pediatric Sedation Outside of the Operating Room, https://doi.org/10.1007/978-3-030-58406-1_4

49

50

nal, and endocrine systems. Some parents may not be familiar with medical terminology or may assume that you are aware of the child’s history; the provider can work around this by describing common problems and/or procedures, pursuing anything that “sounds familiar.” Review any available medical records and contact the primary care provider if possible. Examine previous records in regard to previous problems with airway management, obtaining intravenous access, or prior adverse events related to sedatives-anesthetics. Antenatal history should be reviewed, as maternal medical conditions or complications may affect the neonate adversely. Determine gestational age and conceptional age – premature infants may have pulmonary, cardiovascular, neurologic, gastrointestinal, and hematologic conditions that may lead to decompensation during sedation. Elicit a history of prior sedation-anesthesia and any known adverse reactions, such as marked nausea, vomiting, increased or decreased sensitivity to sedatives or analgesics, and/or prior need for intervention during sedation or unexpected hospitalization after procedures. The complete list of current medications and allergies should be carefully documented. Confirming NPO status is important: children cannot be never be trusted to have fasted. The child and parent should be carefully questioned about any recent intake by mouth, however trivial it may seem.

Physical Examination The initial physical examination provides the sedation practitioner with an opportunity to become familiar with the patient’s baseline physiologic status. Perform a targeted physical examination, including airway assessment, respiratory status, and volume status. Some children will present with a syndrome that the parents do not disclose, either because they assume you are aware or for personal reasons; in these cases, tactfully ask about any special needs. Specific syndromes may be recognized by unusual features, many of which appear as a constellation of associated findings. Inquire as to what extent the child is affected by the syndrome and his current functional status. The final component of a comprehensive pre-anesthetic evaluation is consultation when indicated. It is now common for ASA III and IV patients to seek elective procedural sedation. One must determine if the patient’s medical conditions have been optimized. If not, the procedure must be postponed until this can be accomplished.

When Not to Proceed Barring emergent or life-threatening circumstances, situations arise in which – despite pressure from consultants, providers, and/or families  – the practitioner should forgo

T. Horeczko and M. Mahmoud

sedation outside of the operating room for a more opportune time, setting, or facility. Proper monitoring, rescue equipment, and sufficient staff should be in place. The provider should use sound clinical judgment before proceeding, informed by the patient’s risk for complications and the urgency of the procedure, as well as practical concerns such as the ability to dedicate the necessary time, attention, and human resources to the endeavor.

 hen Referral to General Anesthesia Is W Required Anesthesia providers will never be able to meet the demands for all pediatric procedural sedation. Patient selection for pediatric procedural sedation vs referral to GA is an important issue as last-minute cancelations and inappropriate referrals to pediatric procedural sedation result in family dissatisfaction and unproductive use of sedation team time. Prospective studies examining the characteristics of children who are unable to complete pediatric procedural sedation are required, and guidelines for referral of children to general anesthesia are currently lacking. A recent study approached this issue by combining risk factors for failing sedation with the examination of reasons for referral of children to GA to complete MRI studies. The study showed a significant increase in referrals to GA for an MRI study following implementation of a pediatric procedural sedation guide; however, the authors did not see an impact of their pediatric procedural sedation guide on adverse events or failed sedation outcomes [1]. More multicenter studies are still needed to determine the impact of this sedation workflow, severe adverse event rate, and the appropriateness of referrals to the GA service. The following section is a broad overview that will address specific safety considerations and focused assessments in important special populations.

Fasting Guidelines and Sedation Although the presence of gastric contents theoretically increases the risk of aspiration pneumonia, gastric volumes remain a poor surrogate marker for aspiration risk, and there is no known gastric fluid volume (GFV) that places a particular patient at clinically relevant risk or that eliminates all risk [2]. The traditional teaching is that the risk of aspiration increases with gastric acid volume greater than 0.4  mL/kg and a pH of less than 2.5 [3]. However, if these threshold values were applied, a great number of appropriately fasted patients would be classified as at risk for aspiration. That is, the stomach is rarely completely empty – even in the fasted state  – given ongoing salivary (1  mL/kg/h) and gastric

4  Pre-sedation Assessment

Fig. 4.1  CT of the abdomen without administration of oral contrast in a fasting 2-year-old child in supine position. CT shown in axial (A) plane. Note fluid (labeled “F”) and air (labeled “A”) in a distended stomach. The measured volume of fluid in the stomach was 41.8  ml (3.3 ml/kg). (Courtesy of Mohamed Mahmoud, MD)

(0.6 mL/kg/h) secretions [4]. The provider may expect GFV to be minimal in most fasting patients, but some patients may have large residual GFV despite having followed traditional fasting guidelines (Fig. 4.1). Prolonged fasting in children is still common and is not entirely benign: the fasting child is always at risk for irritability and results in detrimental physiological and metabolic effects including hypoglycemia and/ or hypovolemia. Optimize your patient’s volume and metabolic status before the procedure with the appropriate intravenous fluids if needed. Due to high metabolic needs, an infant should be offered clear fluids until 2 h before sedation. Fasting guidelines regarding clear fluids have been recently changed, and currently, children in several countries are allowed to drink until 1 h prior to anesthesia on the day of surgery [5]. A “true” light breakfast of semisolid content light breakfast 4 h before the procedure has been also advocated by some centers and has been reported to empty from the stomach within 4 h in most children [6]. Re-examining current fasting regimens and national fasting guidelines periodically is recommended, and reducing clear fluid fasting times for elective procedures remains a simple strategy to improve the peri-procedure experience for parents and children. However, providers should be careful with shortening the fasting interval to the point where flexibility to change the order of the procedure is not possible and eventually the ability to run an efficient procedural sedation schedule is difficult. Recently the multidisciplinary International Committee for the Advancement of Procedural Sedation published the first fasting and aspiration prevention recommendations specific to procedural sedation [7]. Green et al. presented a consensus statement that can serve as a resource to sedation

51

providers who perform and oversee procedural sedation. This new consensus-derived algorithm (Fig.  4.2) provides guidance for the management of patients of all ages undergoing procedural sedation. One must be careful with these recommendations since they are not a substitute for clinical judgment. As an example the authors consider bowel obstruction a moderate risk factor for aspiration in the algorithm, and we know that not all bowel obstructions are created equal and procedural sedation may not be acceptable for some scenarios with bowel obstruction. There is a presumption that the relative risk of aspiration is lower during sedation than under general anesthesia and that protective airway reflexes are retained fully during sedation. It is important to note that the progression from mild sedation or analgesia to general anesthesia represents a continuum not easily divided into discrete stages [8]. Anyone receiving moderate or deep sedation should be treated similarly to those receiving general anesthesia because the sedation level can change rapidly and deepen subtly with subsequent impairment of airway reflexes. Although aspiration is a widely feared complication of general anesthesia, fortunately clinically relevant aspiration in modern anesthesia practice is exceptionally rare in pediatrics. The incidence is estimated to be 1  in 10,000 to 10  in 10,000, with the wide reported range likely due to variation in research methodologies, definitions, and reporting sensitivities [9]. In children, pulmonary aspiration is an extremely rare event in, with an incidence of 0.07–0.1% [10]. The recent incidence of severe critical events in pediatric anesthesia (APRICOT) study found an incidence of 9.3/10000  =  00.093%. This study included emergency and unfasted patients as well as elective procedures [11]. In those undergoing general anesthesia, approximately two-thirds of aspiration occurs during manipulation of the airway (endotracheal tube placement and removal) [12]. The multicenter Pediatric Sedation Research Consortium collected data on 49,836 propofol sedations in children: aspiration during sedation occurred 4 times (0.04%) [13]. A retrospective study by Sanborn et al. of 16,467 sedations during imaging procedures in children using chloral hydrate, midazolam, fentanyl, or pentobarbital found 70 (0.4%) respiratory incidents; only 2 patients of 16,467 aspirated (0.012%) [14]. The low incidence of aspiration pneumonia with sedation and anesthesia may be attributed to the fact that the stomach is very distensible and can accommodate a large volume before resting intragastric pressure rises [15]. Intragastric pressure must exceed the barrier pressure of the lower esophageal sphincter (LES) for regurgitation to occur. The barrier pressure of the LES does not appear to be as easily overcome under general anesthesia as is widely believed [15]. The American Society of Anesthesiology’s (ASA) Task Force on Fasting has published consensus guidelines for elective anesthesia: clear fluids, 2  h; breast milk, 4  h; for-

52

T. Horeczko and M. Mahmoud

Pre-sedation assessment - risk factors Negligible risk factors • No risk factors shown to the right

Mild risk factors Patient • Severe systemic disease • Moderate obesity1 • Age 12 months or less • Hiatal hernia Procedure/Sedation • Upper endoscopy • Bronchoscopy • Propofol principal sedative

Moderate risk factors Patient • Severe systemic disease that is a constant threat to life • Severe obesity1, obstructive sleep apnoea • Airway abnormalities2 • Hyperemesis, oesophageal disorders3, bowel obstruction4 Procedure/Sedation • Anticipated need for assisted ventilation or other advanced airway management

Elective procedures Negligible aspiration risk

Clear liquldss Breast milk Food, formula, nonhuman milk

Unrestricted

Moderate aspiration risk (any of the above) Refer for anaesthesia care

Mild aspiration risk (any of the above)

Fasting approximately 2 h 6

Unrestricted

Unrestricted Fasting approximately 2h6

Fasting approximately 2 h 6

Fasting approximately 4 h 6

Fasting approximately 4 h 6

Fasting approximately 6 h 6

Urgent or emergency procedures No delay based on fasting time

No delay based on fasting time

No delay based on fasting time. Anaesthesia care if available; if not consider ketamine as sole sedative agent.

Fig. 4.2  Algorithm linking risk stratification and fasting guidance. Notes: (1) Suggested definitions for moderate obesity are a body mass index (BMI) of 30–39 kg.m-2 in adults or from the 85th up to the 95th BMI percentile based on age/sex in a child, and for severe obesity a BMI of 40  kg.m-2 or higher in an adult or at the 95th percentile or greater in a child. (2) Includes micrognathia, macroglossia, and laryngomalacia; (3) includes gastroparesis, achalasia, atresia, stricture, and tracheoesophageal fistula; (4) includes ileus, pseudo-obstruction,

pyloric stenosis, and intussusception. (5) Clear liquids are generally considered to include water, fruit juices without pulp, clear tea, black coffee, and especially prepared carbohydrate-containing fluids. (6) Fasting intervals are not absolute, with exceptions permissible when the volumes of oral intake are minor, or the fasting time is reasonably close. (Reproduced from Green et  al. [7] with permission from John Wiley and Sons)

mula, 6 h; and solids, 8 h [16]. These guidelines are intended for healthy patients of all ages undergoing elective procedures; they are not intended for patients with coexisting diseases or conditions that may delay gastric emptying such as diabetes, hiatal hernia, gastroesophageal reflux, or bowel obstruction. The ASA acknowledges that there is insufficient evidence to codify preoperative fasting times. In addition, the task force does not offer specific guidance for fasting times for emergency procedures. The ASA pre-procedural fasting guidelines are not applicable to ED procedural sedation. A recent study examined the association between pre-procedural fasting duration and the incidence of sedation-related adverse outcomes in 6183 children and showed that more than 50% did not meet the ASA guidelines for fasting and there were no reported cases of aspiration [17]. When practitioners formulate a plan for sedation for emergency procedures in children who have not

fasted, the risks of sedation and the possibility of aspiration must be balanced against the benefits of performing the procedure emergently. The American College of Emergency Physicians (ACEP) Clinical Policy on Sedation assesses risk based on the nature of the last oral intake and the urgency of the procedure (Fig. 4.3) [18]. In this setting, aspiration has been found to be very rare among patients sedated in an emergency room setting for procedures, regardless of fasting status [19]. There is an ongoing debate regarding the administration of oral contrast for computerized tomography (CT) prior to sedation. The administration of oral contrast less than 2  h before sedation-anesthesia is at odds with elective NPO guidelines and, in theory, would increase the risk of aspiration pneumonia. Sedation practitioners are asked to make an exception to the fasting guidelines and permit the use of enteric contrast material with CT in order to obtain an accu-

4  Pre-sedation Assessment

53

Standard-risk patienta Procedural Urgencyb Emergent Procedure

Urgent Procedure

SemiUrgent

NonUrgent

Nothing

All levels of sedation

All levels of sedation

All levels of sedation

All levels of sedation

Clear liguids only

All levels of sedation

All levels of sedation

Up to and including brief deep sedation

Up to and including extended moderate sedation

Light snack

All levels of sedation

Up to and including brief deep sedation

Up to and including dissociative sedation, non-extended moderate sedation

Heavier snack or meal

All levels of sedation

Up to and including extended moderate sedation

Minimal sedation only

Minimal sedation only Minimal sedation only

Higher-risk patienta Oral intake in the prior 3 hours Nothing Clear liquids only

Light snack

Heavier snack or meal

Procedural Urgencyb Emergent Procedure All levels of sedation

Urgent Procedure

SemiUrgent

NonUrgent

All levels of sedation

All levels of sedation

All levels of sedation

All levels of sedation

Up to and including brief deep sedation

Up to and including extended moderate sedation

Minimal sedation only

All levels of sedation

Up to and including dissociative sedation, non-extended moderate sedation

Minimal sedation only

Minimal sedation only

All levels of sedation

Up to and including dissociative sedation, non-extended moderate sedation

Minimal sedation only

Minimal sedation only

Procedural sedation and an algesia targeted depth and durationc Minimal sedation only

Increasing potential aspiration risk

Oral intake in the prior 3 hours

Dissaciative sedation; berifor intermediate - length moderate sedation

Extended moderate sedation

Brief deep sedation

Intermediate or extended-length deep sedation Brief 20 minutes

Fig. 4.3  Prudent limits of targeted depth of ED procedural sedation. (Reproduced from Green et al. [18] with permission from Elsevier)

rate study. There does not appear to be a perfect resolution to this issue, since waiting several hours after administration of contrast often results in inadequate opacification of the small bowel and a poor study [20]. Small bowel transit time can be as rapid as 15 min and on average is 1 h, 24 min [21]. In one study, in 83% of cases small bowel transit time was less than 2 h [21]. Inadequate opacification of the small bowel can lead to lack of distinction between small bowel loops and fluid collections or masses [20]. At one author’s institution, administration of contrast begins 2 h before and ends 1 h prior to anesthesia-­ sedation. The challenge lies in balancing technical factors governing the image quality of the study with safety concerns related to sedating a child with a potentially full stomach for an elective CT. A recent retrospective chart review concluded that administering an oral contrast material within 2  h of propofol sedation for abdominal CT in children appears to be relatively safe. The data sample, however, was small relative to the reported incidence of aspiration in the literature [22]. Currently we are not aware of any clear consensus among institutions that care for these patients. Some clinicians may choose to perform rapid sequence induction

of general anesthesia with endotracheal intubation, while others may choose deep sedation without definitive airway protection. Others may negotiate with radiologists to have the oral contrast given 2 h before the study or administered through an oral gastric tube after placement of an endotracheal tube [23, 24]. An alternative approach in this scenario or any questionable violation of NPO is to evaluate gastric content with ultrasound (US). US is a noninvasive tool available almost everywhere, and a generation of young sedation providers has grown up with it. It can be utilized routinely in our clinical practice or when we expect an elevated risk due to trauma or short fasting duration [25].

Preparation for and Considerations in Special Populations Asthma and Reactive Airway Disease The child who wheezes presents a common challenge to the sedation practitioner. Transient wheezers are infants whose symptoms are provoked by an active viral respiratory infec-

54

T. Horeczko and M. Mahmoud

tion. These children typically “outgrow” their reactivity within the first few years of life. After the toddler and preschool period, nonatopic wheezers continue to experience wheezing with active viral illnesses, but are not likely to develop lifelong symptoms. Both transient and nonatopic wheezers tend to have mild reactions to the inciting event. Atopic wheezers are equally sensitive to viral illnesses, but often also suffer from allergy, allergic rhinitis, and atopic dermatitis. These children are at highest risk for severe and persistent symptoms exacerbated by a variety of infectious and/or environmental factors [26]. The diagnosis of asthma is difficult to make under the age of 6, since there is a significant overlap with reactive airway disease and pulmonary function tests are problematic in young children. In those with an established diagnosis of asthma, the assessment of symptoms follows a stepwise approach (Table 4.1). In addition to the assessment of severity of symptoms, confirm the overall control of symptoms and what level of therapy the child is currently receiving. It is also helpful to ascertain the responsiveness that the child has shown to previous exacerbations [27]. This is especially important in the planning of procedures that involve airway stimulation or those that would require frequent suctioning. Children with a history of either reactive airway disease or diagnosed asthma are at risk for bronchial hyperreactivity (40% of school-aged children with asthma) [28]. Bronchial hyperreactivity may persist for weeks after an exacerbation. For this reason, a careful history of recent illness, changes in medication, and history of hospitalization are important in all children with a history of wheezing. In general in children with stable and controlled asthma or reactive airway disease, the peri-procedural risk for bronchospasm is low and is not associated with a significant morbidity [29]. A recent prospective study found that patient factors (readily known on pre-procedural assessment) such as active respiratory symptoms, eczema, family history of asthma, rhinitis, or exposure to tobacco smoke were associated with an

increased relative risk of peri-procedural respiratory adverse events such as airway obstruction, oxygen desaturation (2 times/week but 2 times/month

PEF or FEV1 ≥ 80% of predicted or personal best

PEF or FEV1 ≥ 80% of predicted or personal best

Moderate persistent asthma Daily symptoms

Severe persistent asthma Continual symptoms

Exacerbations two or more time/week; may last days Activity causes exacerbations

Frequent exacerbations

>1 time/week PEF or FEV1 > 60% and 150  mEq/L). Nephrogenic DI occurs when the kidney is unable to control plasma osmolality due to a defect in the action of AVP. Medications such as demeclocycline, lithium, amphotericin B, and fluoride [5] and electrolyte abnormalities such as hypokalemia and hypercalcemia [6] are known to cause or precipitate nephrogenic DI. Central DI occurs due to the destruction of the posterior pituitary and eventually lack of AVP production or release. Without treatment, intravascular volume depletion occurs, cardiac stroke volume decreases, and eventually heart rate increases. These patients will have orthostatic hypotension, weak pulses, rapid breathing, and decreased level of consciousness. They may present with seizures if significant hypernatremia is present. A child undergoing procedural sedation should receive his usual morning dose of desmopressin. The sedation provider should pay attention to fluid management in the patient on desmopressin therapy, as some degree of fluid restriction is required. Intravenous fluids (use 5% dextrose-0.9% saline) should total 1  L/m2/24  h to approximate insensible losses and obligate urine output. Oral fluids may be offered once the child is awake.

Mitochondrial Disease Mitochondrial disease (MD) is a group of disorders that arise from defects in the oxidative phosphorylation or electron transport chain involved in the generation of ATP [81]. Primary mitochondrial disorder is caused by deletions in nuclear DNA or mitochondrial DNA.  Secondary disorders are due to mitochondria dysfunction caused by various drugs and by free radicals. The ten most common syndromes associated with mitochondrial disease are Kearns-Sayre syndrome; Leigh syndrome; mitochondrial DNA depletion syndrome; mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS); myoclonic epilepsy with ragged red fibers; neurogastrointestinal encephalomyopathy; neuropathy, ataxia, and retinitis pigmentosa (NARP); and external ophthalmoplegia. There is no definitive treatment for MD, although some patients improve with specific therapies such as coenzyme Q10; those with seizures may respond to a ketogenic diet. Sedating-anesthetizing children with mitochondrial disease may perplex many practitioners. Currently there is no clear evidence-­based guidance in the literature regarding the anesthetic-­ sedative management of these patients. Complicating matters further is the risk of clinical deterioration related to the stress of the procedure itself, unrelated to nature of the anesthetic-­ sedative agents used. It is well known that children with mitochondrial defects may have an increased risk for cardiorespiratory and neurological and

T. Horeczko and M. Mahmoud

metabolic complications from anesthesia-sedation. Any organ may be affected in mitochondrial disease: meticulous individualized pre-sedation assessment is essential. Sedation providers should review and consider obtaining complete blood count, basic metabolic panel, liver function tests, thyroid function tests, sleep studies, and ECG and/or echocardiogram as indicated by the patient’s condition and the associated syndrome. Patients with mitochondrial disease often develop hypoglycemia and lactic acidosis, which can be exacerbated by the stress of the procedure. Hypoglycemia is common: diseased mitochondria cannot keep up with the body’s energy requirements via fatty acid oxidation during stress, which leads to drawing on and rapid depletion of carbohydrate stores. Outpatient procedure should be scheduled as the first case in the morning to minimize fasting intervals, and prolonged procedures require lactate and blood glucose monitoring. Routine use of lactate-free intravenous fluids (such as 5% dextrose-0.9% saline) in all patients with mitochondrial disease undergoing sedation-anesthesia is recommended. This is especially important for infants, as glucose is the major energy supply to the myocardium and hypoglycemia may contribute to myocardial depression. One should be cautious with of patients with mitochondrial disease who are managed with ketogenic diets for control of seizure [82]. Ketogenic diet is high in fat and low in protein carbohydrate and has been used to treat seizures refractory to multiple antiepileptic drugs. The use of ketogenic diet contraindicates the administration of glucose in these patients because the infusion of glucose is associated with a fall in plasma ketones [83]. When these patients receive dextrose, they should be monitored to ensure that they do not develop either hyperglycemia or lactic acidosis. The prevalence of cardiomyopathy in children with mitochondrial disease is reported to be 20% [84, 85]. The severity of MD correlates with the severity of impairment of cardiac function. Cardiac impairment occurs in Barth syndrome, Kearns-Sayre syndrome, ocular myopathy, and MELAS. A pre-procedure baseline ECG is strongly recommended and can be extremely valuable; red flags in the ECG include any form of heart block or prolonged QT. If the screening ECG is abnormal, a cardiology consult is recommended before proceeding with elective sedation-anesthesia in these patients. For those with cardiomyopathy, an echocardiogram within the past year is recommended. There is no absolute contraindication to any particular anesthetic-sedative agent for patients with MD. Many anesthetic agents adversely affect mitochondrial function in vitro, but adverse events in  vivo are only sparsely reported. Furthermore, the anesthetic agents implicated in these cases have been used without incident in many other reports. Opioids, ketamine, midazolam, and dexmedetomidine do not appear to inhibit mitochondrial function. At the present time,

4  Pre-sedation Assessment

there is no need to avoid volatile agents in patients with mitochondrial disease; inhalational anesthetics have been used without ill effects in these children. Keep in mind that patients with mitochondrial disease may have impaired upper way and respiratory response to hypoxia and hypercarbia. Sedative agents should be titrated carefully in order to avoid respiratory depression. Patients with mitochondrial disease may be more susceptible to the effects of lipophilic agents such as propofol. Propofol uncouples oxidative phosphorylation in mitochondria and suppresses ATP production by interfering with the electron transport chain [86]. There are cases in which shortterm use of propofol has resulted in propofol infusion syndrome (acute bradycardia resistant to treatment and progressing to asystole). These patients may have subclinical forms of mitochondrial disease which are uncovered by the infusion of propofol. Single-dose propofol has been used safely in many patients, but the true risk associated with this practice and the safe total dose and duration of infusion are not established. Since there are many sedative-anesthetic alternatives, it is reasonable to avoid the use of propofol infusion in these patients. As in any child with a known myopathy, children with mitochondrial disease are at risk at baseline for rhabdomyolysis. Further, due to abnormal neuromuscular endplates with the subsequent risk of hyperkalemia, a depolarizing agent such as succinylcholine is contraindicated. Note also that patients with mitochondrial disease also exhibit variable sensitivity to the non-depolarizing neuromuscular blocking agents. Many report mitochondrial patients’ experiencing prolonged neuromuscular block with non-depolarizing neuromuscular blocking agents. Careful titration of neuromuscular blocking agents by twitch monitoring and consideration of administration of reversal agents are recommended. To summarize, the most important anesthetic-sedative considerations in these patient are to maintain normoglycemia and normothermia; to avoid any period of hypoxia; to maintain normovolemia; and to avoid metabolic stresses that can lead to or worsen lactic acidosis.

Mucopolysaccharidosis The mucopolysaccharidoses (MPS) are a group of inherited lysosomal storage diseases resulting from defects in glycosaminoglycan degradation. Depending on the specific deficit in the synthesis of proteins involved in glycosaminoglycan degradation, several distinct clinical types include MPS 1 H/S (Hurler/Scheie syndrome); MPS I H (Hurler disease); MPS II (Hunter syndrome); MPS III A, B, C, and D (Sanfilippo syndrome); MPS I S (Scheie syndrome); MPS IV A and B (Morquio syndrome); MPS IX (hyaluronidase

61

deficiency); MPS VII (Sly syndrome); and MPS VI (Maroteaux-Lamy syndrome). Patients with MPS are increasingly presenting for procedures, not only related to the disease (insertion of a port-a-­ cath for treatment or muscle biopsy) but also common elective surgical and diagnostic procedures. These patients are at high risk for anesthesia-related complications especially in remote locations [87]. Fatal perioperative cardiac complications have been reported in these patients [88]. Excessive accumulation of glycogen within the lysosomes of many tissues, including cardiac muscle cells, leads to a progressive cardiomyopathy, valvular abnormalities, and, in the cardiac conduction system, abnormal conduction velocities. Elective procedure under anesthesia/sedation should be postponed until the child has received adequate enzyme replacement therapy and cardiac hypertrophy is reduced to an acceptable level and consequently arrhythmic risk. Glycosaminoglycans infiltrate the connective tissues of the oropharynx and airways, leading to OSA and eventually difficult ventilation and intubation. Glycosaminoglycan accumulation in the atlantoaxial joint may lead to instability which contraindicates cervical extension during intubation and further complicates airway management. Hematopoietic stem cell transplant and enzyme replacement therapy are the preferred treatment for selected types of MPS.  Stem cell transplant has been demonstrated to reduce the incidence of difficult intubation, but enzyme replacement therapy did not. Establishment and maintenance of secure airways have been reported as the most frequently documented anesthetic problem. Older age was associated with an increased risk of difficult intubation, possibly as an effect of the progressive anatomic alterations of MPS [87]. Careful planning, experienced support, and availability of a cart for difficult airway management are important considerations when anesthetizing/sedating these patients.

Multiple Allergies The term “drug allergy” is often misused by clinicians and patients to describe any reaction (proven or perceived) to a medication. The preferred general term is adverse drug reaction, which encompasses the important subcategories. Three clinically relevant subcategories are drug allergy (reaction resulting from an immunologic mechanism), drug intolerance (reaction resulting from non-immunologic and/or unknown reasons), and pseudoallergy (reaction resembling allergy, but with a multifactorial, unknown, or idiosyncratic cause) [89]. It may not be feasible to differentiate the above in the pre-­ sedation assessment [90]. Allergists suggest referring to these events as predictable reactions (drug overdose, side

62

effects, drug-drug interactions) and unpredictable reactions (allergy, intolerance, pseudoallergy). Predictable reactions are often benign and account for approximately 80% of adverse drug reactions. Unpredictable reactions account for the remaining 20%, with allergic or pseudo-allergic reactions comprising 5–10% of adverse drug reactions [89]. Confirming the diagnosis of a drug allergy is not the goal of the pre-sedation assessment; drug provocation testing performed in other settings remains the criterion standard. However, it is important to note that drug allergy is overdiagnosed in children [91]. Although it is prudent to avoid drugs that may have provoked some reaction in the past, when few alternatives remain, the clinician should focus on determining the risk and potential severity of unpredictable reactions during sedation. Type I allergic reactions are immediate and due to drug-specific antibodies; they require prior exposure and sensitization to the drug. Clinical manifestations include urticaria, angioedema, bronchospasm, and/or anaphylaxis. Type II reactions (anti-tissue cytotoxic, e.g., hemolytic anemia or thrombocytopenia) and type III reactions (immune complex, e.g., serum sickness) are readily identified by a history of severe illness or hospitalization. Type IV reactions (the most common) are delayed hypersensitivity reactions evolving over hours to days, and often present with maculopapular exanthems (but may also manifest as eczematous, pustular, or bullous lesions) [89]. Documenting the timing, course of the reaction, and likely inciting drug may help the clinician to understand the safety of the use of the proposed medication during the procedure. Electronic medical records may be a good source of information, as many include entries on when the drug was given and the nature of the reaction [92]. Multiple drug allergy syndrome (MDAS) describes a condition in which the patient experiences allergic or pseudo-­ allergic reactions to related and non-related drugs [93]. Most cases involve urticarial and/or angioedema; however, Stevens-Johnson syndrome and anaphylaxis have been reported. Interestingly, skin testing in these patients may be negative, even after significant clinical manifestations have been documented. These patients typically are older, most are adults, and many have multiple comorbidities and a long past medical history (with many opportunities to become sensitized to many different types of drugs). Information about the pathophysiology of MDAS remains limited, as there is no criterion standard for diagnosis and prospective studies are lacking [90]. Multiple drug intolerance syndrome (MDIS) may be a separate entity from that which is described above. MDIS is defined as a hypersensitivity to three or more drugs which are “chemically, pharmacologically, and immunogenically unrelated, taken on 3 different occasions, and with negative allergy skin tests” [94, 95]. MDIS patients are also typically older; have anxiety, depressive, and/or somatoform symp-

T. Horeczko and M. Mahmoud

toms; and are typically convinced that they are allergic to all drugs. These patients often require allergy and psychiatric consultations as an outpatient [96]. In summary, the pre-sedation assessment should focus on true allergic or pseudo-allergic signs or symptoms associated with a particular drug and the severity of the presentation. When in doubt and feasible, the clinician in this setting may avoid the drug altogether. If there is a conflict or no acceptable alternative, a frank discussion about the risks, benefits, and other possible alternatives is needed.

Muscular Dystrophies The muscular dystrophies (MD) are a group of progressive myopathic disorders characterized by muscle wasting and weakness. The most common are Duchenne and Becker MDs; other types present at different stages in life, with varying degrees of severity and involving different muscle groups: facioscapulohumeral, limb-girdle, distal, oculopharyngeal, and Emery-Dreifuss [97]. The morbidity of the most common, Duchenne and Becker MDs, involves progressive respiratory failure with recurrent lung infections. The disease is characterized by severe proximal muscle weakness, progressive degeneration, and fatty infiltration of the muscles. Symptoms typically appear at the age of 2–6  years; delayed walking beyond 15  months of age is a common initial sign. Affected children never run properly and have difficulty in climbing stairs; only approximately 10% manage to jump with both feet together. Many children require the use of a wheelchair by age 12, and may not live past their 20s [97]. Most MDs involve some degree of cardiomyopathy and all are at risk for heart failure [98]. Other manifestations include pseudohypertrophy of the calves and markedly elevated creatine kinase levels. The progressive nature of the disorder results in restrictive pulmonary disease, multiple contractures, and scoliosis. Due to advances in medical management, many of these patients may now be expected to live into adulthood. The pre-procedure assessment should focus on the child’s overall function (ambulatory or wheelchair) with careful attention to respiratory toilet. The child with disturbed sleep, nightmares, daytime drowsiness, or early-morning headaches may have unrecognized nocturnal hypoventilation. This may be a clue to a recent worsening trajectory of illness and make the child more likely to benefit from noninvasive positive-pressure ventilation during sleep or sedation. Worsening respiratory symptoms may preclude outpatient sedation. Symptoms of dizziness, chest pain, intermittent increased shortness of breath, nausea, and decreased oral intake may be consistent with developing (or worsening) cardiomyopathy. A thorough cardiovascular exam with careful attention

4  Pre-sedation Assessment

to signs of heart failure (hepatic congestion in infants and toddlers, facial and extremity edema in older children, presence of an S3 or precordial heave) is warranted. One-third of these patients have dilated cardiomyopathy by age 14, with nearly all patients developing some degree of cardiomyopathy by age 18. Due to the prevalence of cardiac disorders in these patients, the American Academy of Pediatrics recommends that children with DMD should undergo cardiac evaluation and optimization of cardiovascular status prior to elective anesthesia [99]. While it is important to investigate and optimize cardiovascular status before the elective procedure, these patients can develop complications despite the presence of reassuring pre-procedure tests. Unexplained tachycardia should raise the suspicion of cardiomyopathy. A pre-procedure baseline ECG and potentially an echocardiographic assessment (within a year from the date of the procedure) are recommended to optimize cardiac function and avoid a dysrhythmia. A child with a pre-procedure echocardiogram showing good left ventricular function may not respond adequately to myocardial stress during the procedure. Some children with particular MDs are at higher risk for dysrhythmias and require a prophylactic implantable defibrillator [100]. The severity and progression of skeletal muscular disease may be outpaced by worsening cardiac muscular disease, such as nonischemic cardiomyopathy [101]. Another important concern in these patients is careful evaluation of the airway and respiratory apparatus. These patients may have a difficult airway due to a combination of macroglossia, weak upper respiratory muscles, limited cervical spine mobility, and limited mandibular mobility. DMD is characterized by weakness of the diaphragm, intercostal muscles, and the accessory muscles of respiration, resulting in restrictive pulmonary impairment and a progressive decrease in total lung capacity and vital capacity. For patients with declining respiratory function, it may be necessary to prepare for noninvasive ventilation prior to the procedure. During sedation, patients with MD are at risk for rhabdomyolysis, with subsequent acute renal failure or hyperkalemia. A careful review of the child’s past procedures and outcomes is recommended. Ideally the child is euvolemic prior to the procedure; care should be taken for proper positioning and potentially adjusting positions during long procedures to discourage the development of rhabdomyolysis. Keep in mind that children with MDs are often sensitive to small doses of opioids and sedatives, which may cause a sudden and prolonged apnea [102]. Plan for minimum pre-­ sedation and small titratable aliquots. Controversy exists concerning the role of inhalational anesthetics and succinylcholine in “triggering” rhabdomyolysis or malignant hyperthermia [98, 103–105]. Some experts recommend against their use based on case reports. Many clinicians avoid their use altogether in children with

63

MD. Propofol, dexmedetomidine, and ketamine (among others) have all been used with success in intravenous sedation in these children [98, 106–108]. Nitrous oxide may be considered in children with MD without significant cardiomyopathy or cardiac dysfunction [109].

Musculoskeletal Disorders Children with musculoskeletal disorders may present repeatedly for diagnostic procedures. These children should be managed with sensitivity. Positioning for the procedure can be challenging, especially in those with limb deformities and contractures. Whenever possible, offer the child a position of comfort and minimize focal pressure during sedation. Achondroplasia is the most common nonlethal skeletal dysplasia. There are two causes for this disorder: the child either has a de novo mutation of the fibroblast growth factor receptor 3 gene or inherits the disorder from his parents. These patients have midface hypoplasia, a depressed nasal base, small nasal airways, narrow oropharynx, and upper airway muscle hypotonia, which predisposes them to the development of OSA [110]. They tend to have a large head, a bell-shaped chest, cupping of the ribs, and short arms and legs. Sedative-anesthetic risks in these patients include a challenging airway and increased sensitivity to sedative-­ anesthetic agents. Patients with severe kyphoscoliosis and restrictive lung disease may have baseline hypoxemia and low lung volumes, predisposing them to hypoxemia during sedation. A review of CT scans and MRI of the spine is helpful before sedating these children. Hyperextension of the neck should be avoided, and special consideration should be taken before manipulating the neck due to the possibility of cervical cord compression [111]. The sedation practitioner must be aware of potential complications when sedating a patient with history of significant scoliosis. The primary aim of pre-procedure evaluation is to detect the presence and extent of cardiac or pulmonary compromise. The earlier the age of onset and the more immature the bone growth at the time the process begins, the worse the disease burden. Children with idiopathic scoliosis tend to have less pulmonary embarrassment than children with neuromuscular scoliosis, who may have abnormalities in the central control of breathing and impaired airway reflexes. Poor coordination of laryngeal and pharyngeal muscles may result in abnormal control of secretions and inadequate cough, increasing the risk of aspiration. Respiratory function should be assessed by a thorough history, focusing on functional impairment (exercise tolerance). Physical examination should include a good understanding of vital capacity (review any pulmonary function tests that may be available). If pre-procedure vital capacity is

64

less than 30–35% of predicted, post-procedure ventilation is likely to be required. Cardiac dysfunction may occur in scoliosis from distortion of the mediastinum; patients may develop cor pulmonale from chronic hypoxemia and pulmonary hypertension. Cardiac studies (ECG, echocardiogram) may be performed as indicated. Osteogenesis imperfecta (OI) is an inherited disorder of the connective tissue whose primary manifestation is an increased susceptibility to fractures. Patients usually present with growth retardation, multiple fractures, progressive kyphoscoliosis, vertebral compression, megalocephaly, macroglossia, blue sclera, dentinogenesis imperfecta, bleeding diathesis, and temperature dysregulation. Anesthetic-­sedative challenges in OI include airway anomalies, chronic lung disease (due to kyphoscoliosis, rib fractures, intrinsic pulmonary hypoplasia, and defective lung collagen), coagulation dysfunction, hyperthyroidism, and an increased tendency to develop peri-procedure hyperthermia [112, 113]. Fractures occur from minor trauma and result in severe deformity of the extremities complicating intravenous access and blood pressure cuff placement [112, 113].

Obesity Pediatric obesity has reached alarming epidemic proportions in both industrialized and nonindustrialized countries [114, 115]. This is a major concern as there is a directly proportional relationship between prevalence of comorbidities and childhood obesity [116], and obese children often become obese adults. The prevalence of childhood obesity increased from 4.2% in 1990 to 6.7% in 2010 and is predicted to increase to 9.1% by 2020 [117]. Subsequently, the demand for sedating obese pediatric patients for invasive and noninvasive procedures continues to grow, and sedation providers can expect to be presented frequently with obese patients in NORA locations. Multi-organ pathophysiological changes associated with obesity make comprehensive preoperative evaluation of obese patients a necessity. All obese children must be evaluated for OSA. If the child has OSA, it is important to assess the severity and current management. Cardiology evaluation in the presence of history of multiple episodes of desaturation 10 [127]. The respiratory disturbance index (RDI) is calculated from all respiratory events (including central apnea) occurring in 1 h. AHI and RDI are sometimes used interchangeably, but the bottom line is that they may be used to risk-stratify OSA. Nocturnal oximetry assesses the severity of OSA. Isolated severe desaturation (70” [47]. Although BIS monitor is effective in delineating mild from deep sedation, it is not able to consistently differentiate moderate and deep sedation [42, 48]. Although brain maturation does not occur till puberty, formation of new synapses with changes in the EEG continues after birth and “in the first year of life the EEG is characterized by appearance and disappearance of special patterns and by an increasing synchronization between hemispheres” [49]. Several studies have found BIS to correlate with EEG in children over 1 year of age [50–52] and in older children [53, 54]. However, BIS scores have not correlated well with DOA in younger children less than 2 [55] and specifically in infants [42, 56–58]. In another study, children 75%)

obstruction; expiratory stridor results from lower airway obstruction; and biphasic stridor is present with midtracheal lesions. The evaluation of a patient with stridor begins with a thorough history. The age of onset suggests a cause since laryngotracheomalacia and vocal cord paralysis are usually present at or shortly after birth, whereas cysts or mass lesions develop later in life. Information indicating positions that make the stridor better or worse should be obtained, and placing a patient in a position that allows gravity to aid in reducing obstruction can be of benefit during anesthetic induction. Patients at risk for airway compromise may have either anatomic or physiologic abnormalities that may predispose them. Anatomic abnormalities may cause the oropharyngeal or tracheobronchial airway to be compromised and ventilation to be impaired by small changes in position. The anatomic imbalance between the upper airway soft tissue volume and the craniofacial size contributes to pharyngeal airway obstruction. Pharyngeal size is determined by the soft tissue volume inside the bony enclosure of the mandible. The magnitude of pharyngeal muscle contraction is con-

130

trolled by neural mechanisms, and the interaction between the anatomical balance and neural mechanisms, which are suppressed in sedated patients, determines pharyngeal airway size and patient ability to maintain a patent airway. An anatomic imbalance between the upper airway soft tissue volume and craniofacial size will result in obstruction. Anatomic imbalance may be compensated for by enhanced neural mechanisms that regulate pharyngeal dilator muscles in patients during wakefulness. When neural mechanisms are suppressed during sleep or sedation, relaxation of pharyngeal dilator muscles occurs, and the pharyngeal airway severely narrows [13]. Small changes in function in the setting of normal anatomy may similarly cause inadequate oxygenation. Increasing the distance between the mentum and cervical column will transiently relieve the obstruction. This is achieved by positioning the patient in the sniffing position. Similarly, the sitting position displaces excessive soft tissue outside the bony enclosure through the submandibular space. Laryngomalacia is the most common cause of stridor in infants and is usually benign and self-limited. It occurs during inspiration and is most often due to a long epiglottis that prolapses posteriorly and prominent arytenoid cartilages with redundant aryepiglottic folds that fall into the glottis and obstruct the glottic opening during inspiration (Fig. 8.8). There is little obstruction during exhalation since the supraglottic structures are pushed out of the way during expiration. Intermittent low-pitched inspiratory stridor is the hallmark symptom, which appears during the first 2 weeks of life. Symptoms peak at 6 months of age when they are at their worst and then gradually resolve. Although most children are

Fig. 8.8  Larynx of an infant with laryngomalacia. (Photo courtesy of Reza Rahbar, DMD, MD, Boston Children’s Hospital)

L. R. Ferrari

symptom-free by 18–24 months, the stridor can persist for years. The definitive diagnosis is obtained by direct laryngoscopy and rigid or flexible bronchoscopy. Preliminary examination is usually carried out in the surgeon’s office. A small, flexible fiber-optic bronchoscope is inserted through the nares into the oropharynx, and the movement of the vocal cords is observed [14]. Other etiologies include foreign body aspiration, infection such as croup or laryngotracheobronchitis, edema, or mass lesions such as cyst or tumor. Grunting is a low-pitched sound that results when a patient exhales against a closed glottis and is heard on exhalation. Infants and children often grunt to keep the small airways and alveoli open in an attempt to optimize ventilation and oxygenation. The presence of grunting may be a sign of severe respiratory distress and impending respiratory failure. Underlying causes include pneumonia, acute respiratory distress syndrome, pulmonary edema, congestive heart failure, and abdominal splinting. Wheezing during inspiration or exhalation, or both, indicates intrathoracic obstruction of small airways. It may be a result of intrinsic reactive airways, bronchospasm, or foreign body aspiration. Hypoxemia that is present in the wheezing patient may worsen during administration of sedation. One of the most challenging decisions in caring for children is establishing criteria for cancelation of a procedure in the presence of an upper or lower respiratory infection. Children presenting with symptoms of uncomplicated upper respiratory infection who are afebrile, with clear secretions, and appear otherwise healthy should be able to safely undergo sedation. A history of nocturnal dry cough, wheezing during exercise, and wheezing more than three times in the recent 12 months or a history of present or past eczema may be associated with an increased risk for bronchospasm, desaturation, or airway obstruction [15]. Nasal congestion, purulent sputum production, and a history of reactive airway disease are predictors of adverse respiratory events, and children with these advanced symptoms of upper and potential lower respiratory disease should not undergo sedation [16]. There are many syndromes that have anatomic components related to the airway. A large tongue is associated with Down, Hunter, Hurler, and Beckwith-Wiedemann syndromes. Congenital hypothyroidism and Pompe disease are also associated with a large tongue. Patients with Pierre Robin, Treacher Collins, and Goldenhar syndromes, as well as children with congenital hemifacial microsomia, have micrognathia, high arched palate, and a potential to have early airway obstruction when sedated. Children with tonsillar hypertrophy are at risk for mechanical airway obstruction due to large tonsils occupying a greater portion of the oropharyngeal airway than normal-sized tonsils. Former premature infants are at risk for untoward respiratory events during sedation. There is a more gradual slope of the CO2 response curve in the preterm infant, which

8  The Pediatric Airway: Anatomy, Challenges, and Solutions

predisposes this group of patients to apnea. All neonates exhibit periodic breathing, which is manifested as interrupted ventilation by self-corrected short periods of apnea without desaturation or bradycardia [17]. This tendency diminishes by 45 weeks postconceptual age. Apnea of prematurity and postanesthetic apnea are predominantly central in origin, with about 10% due to mechanical obstruction. The response to airway obstruction with apnea is common in infants with periodic breathing and decreases with increasing postnatal age. In the sedated neonate and former premature infant, benign periodic breathing may evolve into frank apnea, which must be managed by stimulation or assisted ventilation. To detect postanesthetic or post-sedation apneic events, it is suggested that infants whose age is under 56 weeks postconception be monitored for 24 h after the procedure [18]. Conditions that interfere with the integrity of the laryngeal inlet or upper larynx may impair effective ventilation as a result of partial or complete airway obstruction. Upper respiratory infections cause increased secretions, which may occlude the larynx in addition to the inflammatory response that can compromise the internal diameter of the laryngeal inlet. Laryngotracheobronchitis or croup also decreases the internal laryngeal diameter and produces the same clinical outcome. The incidence of epiglottitis has decreased dramatically in the past decade but may still be encountered. These patients have not only inflammation of the epiglottis but edema of the surrounding structures, which severely restricts the size of the larynx and encroaches on the area for ventilation to occur. Patients who have sustained airway trauma or thermal injury should be considered in this category as well. Children who have experienced prolonged intubation may have decreased laryngeal inlet diameter as a result of fibrosis from congenital or acquired subglottic stenosis (Figs. 8.9 and 8.10). Any agent that will decrease the pharyngeal muscle tone and rate and depth of respiration in this setting should be given with extreme caution and warrants vigilance. Other conditions that restrict the laryngeal inlet are subglottic stenosis, laryngeal cysts, and papillomatosis. There is a similar concern for narrowing and compromise of the larynx from external factors. Goiter or other tumors of the neck that are extrinsic to the larynx may cause compression and functional restriction to ventilation. Children with arthrogryposis or congenital abnormalities in which the neck is fused may have difficulty with positioning and subsequent ventilation when airway function is depressed during sedation. Children with an anterior mediastinal mass are at significant risk for airway compromise during sedation due to compression of the intrathoracic larynx (Figs.  8.11 and 8.12). Although lymphomas constitute the largest group of masses that arise in the anterior mediastinum, other masses

131

Fig. 8.9  Larynx of an infant with congenital subglottic stenosis. (Reza Rahbar, DMD, MD, Boston Children’s Hospital)

Fig. 8.10  Larynx of an infant with acquired post-intubation subglottic stenosis. (Photo courtesy of Reza Rahbar, DMD, MD, Boston Children’s Hospital)

that may present in this location include teratomas, cystic hygromas, thymomas, hemangiomas, sarcomas, desmoid tumors, pericardial cysts, and diaphragmatic hernias of the Morgagni type. To understand the pathophysiology of the anterior mediastinum, it is important to be familiar with the anatomy. The mediastinum is defined as the extrapleural space in the thorax that is bounded anteriorly by the sternum, posteriorly by the thoracic vertebrae, superiorly by the thoracic inlet, and inferiorly by the diaphragm. Structures contained within the mediastinum that may undergo compression from an ­enlarging mass are the trachea and the main stem bronchi, superior vena cava, aortic arch, main pulmonary artery, and a portion of the heart itself.

132

Fig. 8.11  A 20-month-old male with a large anterior mediastinal mass

L. R. Ferrari

to some degree of airway compromise that may worsen when positive intrathoracic pressure is generated. Cardiovascular symptoms result from compression of the aortic and pulmonary vessels, as well as the right atrium and right ventricle. This can lead to both hypotension secondary to inadequate cardiac filling and restricted pulmonary blood flow resulting in poor oxygenation despite adequate ventilation. Findings referable to the cardiovascular system include fatigue, headache, hypotension or pallor in the supine position, a feeling of light-headedness, superior vena cava syndrome (facial edema, cyanosis, jugular venous distension), and the appearance of a new murmur, especially in the area of the pulmonary valve. It is essential that the practitioner search for these signs and symptoms when interviewing and examining patients with mediastinal masses in an attempt to ascertain the degree of respiratory and cardiovascular compromise present. Patients with minimal symptoms can have catastrophic events if subtle indicators are overlooked. Improvement of these physiologic changes is often quickly achieved by moving the patient into a sitting or left lateral position.

Sleep-Disordered Breathing

Fig. 8.12  CT scan illustrating a large anterior mediastinal mass compressing the lung and causing tracheal deviation

Patients with anterior mediastinal masses may present with varied signs and symptoms referable to both the cardiovascular and respiratory systems and are directly related to the location and size of the mass, as well as the degree of compression of surrounding structures. The most commonly observed respiratory symptom is cough, especially in the supine position, which results from anterior compression of the trachea. Infants younger than 2 years of age are more likely to experience wheezing as a sign of tracheal compression, whereas children older than 2  years of age usually present with malaise, cough, fever, and neck mass. Other respiratory findings in patients of all ages include tachypnea, dyspnea, stridor, retractions, decreased breath sounds, and cyanosis on crying, all of which should alert the practitioner

Sleep-disordered breathing (SDB) is a spectrum of disorders ranging from primary snoring to obstructive sleep apnea syndrome (OSAS). The mildest form of SDB is primary snoring, which is noisy breathing without clinical manifestations and occurs in 20% of normal children [19]. Although SDB affects 10% of the population, only 1–4% will progress to OSAS.  Obstructive sleep apnea syndrome is a disorder of airway obstruction with multisystem implications and associated complications. It affects children from infancy to adulthood and is responsible for behavioral, cognitive, and growth impairment as well as cardiovascular and perioperative respiratory morbidity and mortality [20]. OSAS is associated commonly with comorbid conditions, including obesity and asthma, and is characterized by periodic, partial, or complete obstruction of the upper airway during sleep. Airway obstruction is characterized by an anatomic imbalance between the upper airway soft tissue volume and craniofacial size. Suppression of pharyngeal dilator muscles during sleep and anesthesia occurs in the patient with obstructive sleep apnea, as opposed to patients who are just noisy breathers or have mild to moderate snoring. Repetitive arousal from sleep to restore airway patency is a common feature as are episodic sleep-associated oxygen desaturation, hypercarbia, and cardiac dysfunction as a result of airway obstruction. Individuals who experience obstruction during sleep may have snoring loud enough to be heard through closed doors or observed pauses in breathing during sleep. They may awaken from sleep with a choking

8  The Pediatric Airway: Anatomy, Challenges, and Solutions

sensation. Parents report restless sleep in affected children and frequent somnolence or fatigue while awake despite adequate sleep hours. These children fall asleep easily in non-­stimulating environments and are difficult to arouse at their usual awakening time. Type 1 OSAS is characterized by lymphoid hyperplasia without obesity, whereas type 2 OSAS patients are obese with minimal lymphoid hyperplasia. Approximately, 10% of OSAS is present in preschool and school-aged children and is thought to decline after 9 years of age [4]. Obesity changes craniofacial anthropometric characteristics; therefore body mass index of 95% for age or greater is a predisposing physical characteristic that increases the risk of developing OSAS. Children with craniofacial abnormalities including a small maxilla and mandible, large tongue for given mandibular size, and thick neck have a similar increased risk. Many of these children have syndromes that are associated with additional comorbidities. Anatomic nasal obstruction and Class 4 touching tonsils reduce oropharyngeal cross-sectional area, which constitutes an additional risk. Pharyngeal size is determined by the soft tissue volume inside the bony enclosure of the mandible, and an anatomic imbalance between the upper airway soft tissue volume and craniofacial size will result in obstruction. The magnitude of pharyngeal muscle contraction is controlled by neural mechanisms, and the interaction between the anatomical balance and neural mechanisms determines pharyngeal airway size. Increased neural mechanisms can compensate for the anatomical imbalance in obstructive sleep apnea patients during wakefulness. When the neural mechanisms controlling pharyngeal dilator muscles are suppressed during sleep or anesthesia (as is present in non-­ OSAS patients), the pharyngeal airway severely narrows because of the anatomical imbalance. There is additional decrease in ventilator response and impairment of the arousal response. Craniofacial morphology may influence the severity of obstruction in boys more than girls [21]. Increasing bony enclosure size will provide relief of airway obstruction. This is only accomplished surgically by mandibular advancement. Increasing the distance between the mentum and cervical column by positioning will transiently relieve the obstruction as long as the sniffing position is maintained. Similarly, the sitting position displaces excessive soft tissue outside the bony enclosure through the submandibular space. The long-term effects of OSAS are not limited to the airway. These children have other systemic comorbidities. Increased body mass index and obesity may lead to increased cognitive vulnerability as illustrated by the increased frequency of hyperactivity and increased levels of C-reactive protein. The duration of OSA has no relation to reversibility of neurobehavioral impairment since many believe that episodic hypoxia alters the neurochemical substrate of the prefrontal cortex causing neuronal cell loss. Metabolic

133

syndrome consists of insulin resistance, dyslipidemia, and hypertension. It is felt that OSAS is a risk factor for metabolic syndrome in obese children but not in nonobese patients. Cardiovascular and hemodynamic comorbidities are more common in OSAS patients. These consist of altered regulation of blood pressure as well as alterations in sympathetic activity and reactivity. Also present are endothelial dysfunction and initiation and propagation of inflammatory response facilitated by increases in levels of C-reactive protein [22, 23]. Systemic inflammation using interleukins as a marker is a component of OSAS in both obese and nonobese children and is reversed after tonsillectomy. Systemic hypertension, changes in left ventricular geometry, and intermittent hypoxia leading to pulmonary artery hypertension are well-­described comorbidities present in patients with OSAS. The mainstay of OSAS management is surgical removal of tonsils and adenoids, which carries an 85% success rate in resolving OSAS. Recurrence may occur in children with craniofacial abnormalities and in others, and if surgical intervention does not resolve the problem, continuous positive airway pressure (CPAP) at night is the next treatment modality. Many of these children, however, may present for imaging or require sedation prior to removal of the tonsils or adenoids. For patients undergoing sedation, the preoperative evaluation begins with the history (refer to Chap. 4). Questions to ask parents include the presence of difficulty breathing during sleep, snoring, gasping, retractions, apnea during sleep, sweating during sleep, restless sleep or behavioral problems, and/or somnolence during the day, feeling un-refreshed after awakening [24, 25]. A positive finding of any of the aforementioned characteristics should alert the practitioner to the possibility of some degree of OSAS [26]. The Snoring, Trouble Breathing, and Un-Refreshed (STBUR) questionnaire is a screening tool for pediatric sleep-disordered breathing and risk for perioperative respiratory adverse events in children (Table  8.1). The Snoring, Trouble Breathing, and Un-Refreshed questionnaire identified patients at higher risk for prolonged phase 1 recovery, oxygen therapy requirement, and escalation of care. The questionnaire’s high negaTable 8.1   The STBUR questionnaire for predicting perioperative respiratory adverse events in children at risk for sleep‐disordered breathing Symptom items included in the STBUR questionnaire (STBUR: Snoring, Trouble Breathing, Un-Refreshed) While sleeping, does your child:  Snore more than half the time?  Snore loudly?  Have trouble breathing or struggle to breathe? Have you ever seen your child stop breathing during the night? Does your child wake up feeling unrefreshed in the morning? Reprinted with permission from: Tait et al. [28] with permission from John Wiley and Sons

134

tive predictive value and specificity may make it useful as a screening tool to identify patients at low risk for prolonged stay in PACU [27, 28]. Specific attention should be paid to the frequency of tonsillar infection, recent upper respiratory infections, SDB, and cardiovascular abnormalities. The physical exam should include observation of audible respiration, mouth breathing, nasal quality of speech, chest retractions, long facies, retrognathic mandible, and inspection of tonsillar size. Auscultation should be specifically directed to detect wheezing and stridor. Polysomnography (PSG), otherwise known as the sleep study, is the gold standard for diagnosis of OSAS. A sleep study is suggested to direct the postoperative or postprocedural disposition. Drug-induced sleep endoscopy is becoming an increasingly common technique for identifying anatomical sites contributing to obstructive sleep apnea. This technique consists of the direct examination of the upper airway using flexible endoscopy under deep sedation to identify specific anatomic sites of obstruction [29–31]. It is essential in patients with comorbidities and high-risk features such as morbid obesity, craniofacial abnormalities, neuromuscular disorders, cor pulmonale, systemic hypertension, difficulty breathing during sleep, growth impairment due to chronic obstructed breathing, and a history of severe prematurity [32]. Obesity changes craniofacial anthropometric characteristics, and a body mass index of 95% for age or greater is a risk factor for OSA, which should be quantified by PSG. Craniofacial abnormalities that specifically include small maxilla and mandible, large tongue for given mandible size, and thick neck similarly should be evaluated by sleep study. Despite this, most patients do not have this examination prior to surgery. It is expensive, time-consuming, and unavailable in some medical centers. The nadir of oxygen saturation and respiratory disturbance index (RDI), which is the number of apneic episodes per hour, are measured during PSG. Apnea is defined as decreases in airflow greater than 90% for two breaths or more. Hypopnea is defined as decreases in airflow greater than 50% coupled with 3% decrease in oxygen saturation or electroencephalogram (EEG) arousal. An RDI of two or more is necessary for the diagnosis of OSAS. Mild OSAS is defined as RDI of 5–10 events, moderate 10–20 events, and severe 20–30 events. The STOP-BANG questionnaire has been in use in the adult population since 2009 to predict the presence of OSA in the absence of a sleep study [26]. It is comprised of eight questions designed to predict moderate to severe OSA.  Although it has good predictive value for alerting practitioners to adults with OSA, it is not a good predictor of sedation-related adverse events (SRAE) in children [33]. Although the presence of OSA does not seem to be a risk factor for hypoxia in adults undergoing moderate sedation, this has not been demonstrated in the pediatric population [34].

L. R. Ferrari

When sedation without a secured airway is planned, it is imperative that the level of consciousness, adequacy of ventilation, and oxygenation be continuously monitored and the risk of apnea be evaluated. Patients exposed to recurrent hypoxia exhibit an altered response to narcotics, which is manifested by decreases in minute ventilation, respiratory frequency, and tidal volume. It is therefore suggested that no sedative premedication be administered to OSAS patients prior to a general anesthetic and narcotics be administered in incremental doses, beginning with one-half the recommended dose, until adequacy of ventilation and respiration is determined. Patients with OSAS who are given the same dose of narcotic as non-OSAS patients have a very high risk of serious respiratory compromise [35, 36]. Similarly, patients should not be discharged until fully awake and breathing at a baseline rate and depth. The supraglottic obstruction secondary to decreased muscle tone may contribute to desaturation. Children who have increased severity of OSAS, low weight, and age under 3 years exhibit a higher rate of complications [37]. They are more likely to require supplemental oxygen, the use of an oral airway, and assisted ventilation. Slow return of upper airway tone may lead to desaturation and laryngospasm on emergence, especially in those patients who are known to have an RDI greater than 30. There is no agreement on the specific criteria that preselect an elective OSAS patient for admission and monitoring postprocedure [38]. Inclusive characteristics may include the following: PSG-­ proven OSAS with RDI >40, RDI >20 plus either desaturation