Genetic disorders and the fetus : diagnosis, prevention, and treatment [Eighth ed.] 9781119676935, 1119676932

896 92 22MB

English Pages [1395] Year 2021

Report DMCA / Copyright

DOWNLOAD FILE

Polecaj historie

Genetic disorders and the fetus : diagnosis, prevention, and treatment [Eighth ed.]
 9781119676935, 1119676932

Table of contents :
Cover
Title Page
Copyright
Contents
Preface
Acknowledgments
Contributors
Chapter 1 Genetic Counseling: Preconception, Prenatal, and Perinatal
The burden of genetic disorders and congenital malformations
Incidence and prevalence of genetic disorders and congenital malformations
Congenital malformations and infant morbidity and mortality
Down syndrome
The goal and purpose of prenatal diagnosis
Prerequisites for genetic counseling
Knowledge of disease
Expertise in genetic counseling
Ability to communicate
Knowledge of ancillary needs
Empathy
Sensitivity to parental guilt
Guiding principles for genetic counseling
Accurate diagnosis
Nondirective counseling
Concern for the individual
Truth in counseling
Confidentiality and trust
Timing of genetic counseling
Parental counseling
Counselee education
Duty to recontact
Do no harm
Duty to warn
Preconception genetic counseling
Indications for preconception genetic counseling
Identification of preconception options
Genetic counseling as a prelude to prenatal diagnosis
Informed consent
Presymptomatic or predictive testing
Expansion mutations and anticipation
Disorders with anticipation
Disorders with suspected anticipation
Imprinting and uniparental disomy
Genotype–phenotype associations
Somatic mosaicism
Genetic counseling when the fetus is affected
Decision making
Elective abortion: decision and sequel
Testing the other children
Perinatal genetic counseling
Family matters
The surviving children
The efficacy of genetic counseling
References
Chapter 2 Preimplantation Genetic Testing
Approaches to preimplantation genetic testing
Polar body‐based preimplantation genetic testing
Preimplantation genetic testing based on embryo biopsy
Prospects for noninvasive preimplantation genetic testing
Preimplantation genetic analysis
Single‐gene disorders
De novo mutations
Late‐onset disorders
HLA typing
Chromosomal disorders
Ethical and legal issues
Conclusion
References
Chapter 3 Amniotic Fluid Constituents, Cell Culture, and Neural Tube Defects
Introduction
Amniotic fluid
Formation and circulation
Volume
Origin
Biochemical and other characteristics of amniotic fluid
Cell‐free DNA and RNA
Proteins
Proteomics
Lipids
Enzymes
Amino acids
Disaccharidases
Miscellaneous biochemical constituents and other characteristics of amniotic fluid
Blood group substances
Immunoglobulins
Antibacterial activity of amniotic fluid
Bacteriostatic effect
Isolation of infectious agents
Hormones
Drugs/toxicants
Amniotic fluid cell culture
Alternatives to cell culture and metaphase karyotype analysis
Amniotic fluid cell types
Cell culture and cell harvest
Enhancement of amniotic fluid cell growth
Testing and handling fetal bovine serum
Defined growth factor supplements
Culture failure
Safety in the laboratory
Mesenchymal stem cells in amniotic fluid
References
Chapter 4 Molecular Aspects of Placental Development
Overview
Placental structure
Placental development and function
Implantation
Angiogenesis
Nutrient delivery
Immune function
Placental insufficiency
Fetal growth restriction
Genetic causes of fetal growth restriction
Developmental considerations in confined placental mosaicism
Imprinting and fetal growth restriction
Preeclampsia
Early diagnosis of preeclampsia
Genetics of preeclampsia
Genetic findings associated with molar changes in the placenta
Complete hydatidiform mole
Partial hydatidiform mole
Placental mesenchymal dysplasia
DNA methylation studies in the placenta and their clinical application
Epigenetic studies in the placenta and environment
The placenta as a predictor of child health
Further considerations
References
Chapter 5 Fetal Origins of Adult Health and Disease
Introduction
Epigenetics and programming
Energy‐balance programming: under‐ and overnutrition
Environmental toxins
Maternal stress and anxiety
Glucocorticoids and prematurity
Organ‐specific programming effects
Appetite and adiposity
Hepatic programming
Pancreas programming
Cardiac programming
Bone programming
Brain programming
Renal programming
Immune system programming
Endocrine programming
Sexuality programming
Conclusion
References
Chapter 6 Maternal Serum Screening for Chromosomal Abnormalities and Neural Tube Defects
Chromosomal abnormalities
Neural tube defects
Screening and prenatal diagnosis
Widely used markers
Additional markers
Marker distributions in Down syndrome, neural tube defect, and unaffected pregnancies
Risk screening for Down syndrome
Age‐specific Down syndrome risk at term
Down syndrome risk at the time of the test
Down syndrome likelihood ratios
Modeling performance of Down syndrome screening
Established multimarker Down syndrome policies
Model performance of neural tube defect screening
Prospective confirmation of the Down syndrome model
Further multimarker Down syndrome strategies
First‐trimester Contingent test
Additional first‐trimester ultrasound markers
Additional first‐trimester serum markers
First‐trimester Triple and Quad tests
Second‐trimester Combined test
Genetic sonogram
Repeat measures and highly correlated markers
Two‐sample Combined test
Ultrasound screening for OSB
Second‐trimester lemon and banana signs
Second‐trimester anomaly scan
First‐trimester anomaly scan
First‐trimester screening
Other Down syndrome markers
A disintegrin and metalloprotease 12s (ADAM12s)
Pregnancy‐specific glycoprotein‐1
Urinary human chorionic gonadotropin species
Serum invasive trophoblast antigen
Serum thyroid‐stimulating hormone
Clinical factors
Maternal age
Previous affected pregnancy
Twins
Assisted reproduction
Maternal diabetes
Renal transplant
Previous false positive
Smoking
Ethnicity
Maternal weight
Other factors
Edwards syndrome (trisomy 18)
Other conditions associated with altered markers
Other chromosome abnormalities
X‐linked ichthyosis
Smith–Lemli–Opitz syndrome
Cornelia de Lange syndrome
Abdominal wall defects
Cardiac abnormalities
Moles and placental mesenchymal dysplasia
Fetal demise
Adverse maternal–fetal complications of pregnancy
Planning a program
Conclusion
Acknowledgments
References
Chapter 7 Noninvasive Screening for Aneuploidy Using Cell‐Free Placental DNA
Introduction
Cell‐free DNA
Performance of cell‐free DNA screening
Sex chromosome aneuploidy
Cell‐free DNA screening approaches
Cell‐free DNA test failures
False‐positive cell‐free DNA results and incidental findings
Maternal malignancy
Vanishing twins
False‐negative cell‐free DNA results
Cell‐free DNA screening for microdeletion syndromes
Genome‐wide cell‐free DNA screening
Pretest counseling
Post‐test screening
Comparison of cell‐free DNA screening to traditional screening
Cost‐effectiveness
Contingent screening
Cell‐free DNA screening following a positive traditional screening test
Use of prenatal ultrasound in the setting of cell‐free DNA
First trimester
Ultrasound soft markers and cell‐free DNA screening
Discordance between fetal sex on ultrasound and cell‐free DNA screening
Multiple gestations
Cell‐based noninvasive prenatal testing
Conclusion
References
Chapter 8 Noninvasive Prenatal Diagnosis and Screening for Monogenic Disorders Using Cell‐Free DNA
Introduction
Biology and characteristics of cell‐free DNA in maternal blood
Origin of fetal cell‐free DNA
Importance of the fetal fraction
General approaches for testing of single‐gene disorders by fetal cell‐free DNA analysis
Identification of de novo or paternally inherited variants
Identification of maternally inherited variants
Laboratory techniques used for cfDNA‐based single‐gene disorder analysis
Limitations of cfDNA‐based detection of single‐gene disorders
Current status of noninvasive single‐gene testing by cell‐free DNA analysis
Overview
Noninvasive fetal sex determination
Fetal RHD and other blood group genotyping
Noninvasive prenatal diagnosis of monogenic disorders
Skeletal dysplasias
Duchenne and Becker muscular dystrophy
Cystic fibrosis
Spinal muscular atrophy
Congenital adrenal hyperplasia
Hemoglobinopathies
Noninvasive prenatal screening using panels of single‐gene disorders by cell‐free DNA analysis
Clinical implementation: ethical and social issues
Summary and future directions
References
Chapter 9 Amniocentesis, Chorionic Villus Sampling, and Fetal Blood Sampling
Introduction
Amniocentesis
Prerequisites
Timing
Technique
Ultrasound guidance during amniocentesis
Amniocentesis in multiple gestations
Rh isoimmunization in amniocentesis
Significance of amniotic fluid discoloration
Safety of genetic amniocentesis
Pregnancy losses
Early amniocentesis
Third‐trimester amniocentesis
Chorionic villus sampling
Technique of chorionic villus sampling
Complications of chorionic villus sampling
Safety of chorionic villus sampling in multiple pregnancies
Reliability of results from chorionic villus sampling
Fetal abnormalities following chorionic villus sampling
Fetal blood sampling
Fetal hematologic disorders
Fetal infection
Fetal therapy
Technique of fetal blood sampling
Safety of fetal blood sampling
Fetal blood sampling in multifetal pregnancies
Fetal blood sampling in fetuses with single umbilical arteries
First‐trimester fetal blood sampling
Cardiocentesis
References
Chapter 10 Prenatal Diagnosis of Neural Tube Defects
Biology of α‐fetoprotein
Amniotic fluid α‐fetoprotein
Multiple pregnancy
Causes of elevated (or low) levels of AFAFP in the absence of NTDs
Likely mechanism/condition
Leakage through skin
Urinary tract leakage
Leakage of placental origin
Leakage of pulmonary origin
Reduced intestinal AFP clearance or leakage
Unknown site of “leakage”
Problems and pitfalls
Amniotic fluid acetylcholinesterase
Experience with AFAChE
Recommendations for prenatal diagnosis of NTDs using AFAFP and AChE assays
Other techniques to detect neural tube defects
Primary prevention of neural tube defects
Genetic counseling
Nutritional supplementation
Complications and life expectancy
References
Additional references
Chapter 11 Prenatal Diagnosis of Chromosomal Abnormalities through Chorionic Villus Sampling and Amniocentesis
The incidence of chromosomal abnormalities detected by conventional cytogenetics
Data from livebirths
Data from adult biobanks
Data from amniocentesis
Data from chorionic villus sampling
Data from spontaneous abortuses
Data from induced abortuses
Data from stillbirths
Indications for prenatal cytogenetic diagnosis
Noninvasive prenatal testing and trisomy 21, trisomy 18, and trisomy 13
Noninvasive prenatal testing and sex chromosome abnormalities
Noninvasive prenatal testing and microdeletion syndromes
Genome‐wide noninvasive prenatal testing
Noninvasive prenatal testing and low fetal fraction
The first‐trimester Combined test
Second‐trimester maternal serum screening
Elevated maternal serum ?‐fetoprotein
Abnormal ultrasound findings
The “genetic sonogram” or “anomaly scan”
Advanced maternal age
Advanced paternal age
Multiple gestation pregnancy
Carrier of a balanced structural rearrangement
Previous child with trisomy
Previous child with a de novo unbalanced rearrangement: isochromosome 21q and others
Genetic variation in folate metabolism and previous child with a neural tube defect
History of repeated fetal losses: parents' karyotype unknown
History of fetal loss: fetal karyotype considerations
Fetal demise, current pregnancy
Male or female subfertility, cytogenetic causes
Reduced ovarian complement
Abnormal parental karyotype (other than a balanced structural rearrangement)
Prenatal sex determination for X‐linked disorders
Prenatal diagnosis for a nonchromosomal disorder
Miscellaneous
Interpretation issues: chromosome mosaicism and pseudomosaicism
General considerations
Diagnosing mosaicism in chorionic villus sampling
Diagnosing mosaicism in amniotic fluid cell cultures
Mosaicism involving gain of an autosome: data for individual chromosomes
Autosomal monosomy mosaicism
Mosaicism involving an autosomal structural abnormality (excluding supernumerary marker chromosomes) in CVS
Mosaicism involving an autosomal structural abnormality (excluding supernumerary and marker chromosomes) in AFC
Sex chromosome mosaicism in chorionic villus sampling
Sex chromosome mosaicism in amniotic fluid cells
Other types of mosaicism
Guidelines for the diagnosis of mosaicism
Genetic counseling and chromosome mosaicism
Interpretation issues: chromosome rearrangements
Familial structural rearrangements
De novo structural rearrangements
Uniparental disomy in familial and de novo rearrangements
Summary conclusions and recommendations for chromosome rearrangements
Interpretation issues: chromosome polymorphisms, common inversions, and other structural variations
Polymorphisms of chromosomes 1, 9, 16, and Y
Polymorphisms of acrocentric chromosomes
Polymorphism of other chromosomes, “common” inversions, and translocations
Summary conclusions and recommendations for polymorphisms and other variations
Interpretation issues: maternal cell contamination
Maternal cell contamination in chorionic villus sampling
Maternal cell contamination in amniotic fluid cell culture
Factors affecting diagnostic success rate and accuracy
Twin pregnancy
Mycoplasma contamination of cell cultures
Toxic syringes or tubes
Other causes of culture failure
Technical standards for prenatal cytogenetics laboratories
Error rates in prenatal cytogenetic diagnosis
Discordance between karyotyping and molecular genetic testing
Conclusion
Acknowledgments
References
Chapter 12 Prenatal Diagnosis of Sex Chromosome Abnormalities
Incidence
Ascertainment bias
Patterns of inheritance
Prenatal diagnosis
Turner syndrome
Diagnosis and management
Cognitive/psychologic development
Karyotype variations
Prenatal counseling for Turner syndrome
45,X mosaicism
Klinefelter syndrome
Clinical features and management
Cognitive/psychologic development
Prenatal counseling for 47,XXY
47,X,i(Xq),Y
47,XXY mosaicism
48,XXYY
48,XXXY
49,XXXXY
49,XXXYY
Triple X and poly‐X syndromes
Clinical features and medical management
Cognitive/psychologic development
Prenatal counseling for 47,XXX
47,XXX mosaicism
48,XXXX
49,XXXXX
47,XYY males
Historical perspective
Clinical features and medical management
Cognitive/psychologic development
Prenatal counseling for 47,XYY
46,XY/47,XYY mosaicism
Polysomy Y karyotypes
48,XYYY
49,XYYYY
49,XXYYY
Structural abnormalities of the X chromosome
Xp deletions: del(Xp) or Xp2
Xq deletions: del(Xq) or Xq2
Xp duplications: dup(Xp)
Xq duplications: dup(Xq)
Isochromosome Xp: i(Xp)
Isochromosome Xq: i(Xq)
Marker X
Inversion X: inv(X)
X;autosome translocations
X;X translocations
Structural abnormalities of the Y chromosome
Yp deletions: del(Yp)
Yq deletions: del(Yq)
Isochromosome Yp: i(Yp)
Isochromosome Yq: i(Yq)
Isodicentric Yp: idic(Yp)
Isodicentric Yq: idic(Yq)
Ring Y: r(Y)
Marker Y: mar(Y)
Inversion Y: inv(Y)
Satellited Yq: Yqs
Y;autosome translocations
X;Y translocations
Y;Y translocations
Disorders of sex development
46,XX males
45,X males
47,XXX males
46,XY females
Other sex reversal syndromes
Ovotesticular disorders of sex development
Conclusion
References
Chapter 13 Prenatal Diagnosis of Chromosomal Abnormalities: From Karyotype to Microarray
The study and impact of chromosome abnormalities in humans
Traditional cytogenetic testing: analysis of the G‐banded metaphase
Incidence and spectrum of chromosome abnormalities observed in prenatal diagnosis
Rapid identification of the common aneuploidies
Fluorescence in situ hybridization
Quantitative fluorescence polymerase chain reaction
Multiplex ligation‐dependent probe amplification
Chromosome microarray analysis adds diagnostic yield over karyotyping and rapid aneuploidy techniques
Types of microarrays
Array comparative hybridization
Single‐nucleotide polymorphisms arrays
Microarray design for clinical testing
Interpreting and reporting of CMA results
Cytogenomic tools and tips for interpreting CNVs
Factors affecting CMA diagnostic yield
Number of probes on the array and CNV size cutoff
Impact on variants of uncertain significance
The benefit of SNPs
CMA in routine pregnancies
CMA in pregnancies with ultrasound anomalies
CMA versus karyotyping: additional points to consider
CMA and genetic counseling
Conclusion
References
Chapter 14 Molecular Genetics and Prenatal Diagnosis
Diagnostic methods: use, limitations, and pitfalls
Source of DNA for analysis
Methods of analysis
Clinical databases
In silico tools
Carrier detection
Presymptomatic/predictive DNA tests
Mutation detection
Clinical caveats, cautions, limitations, and pitfalls
Dynamic mutations and anticipation
Mosaicism
Imprinting and uniparental disomy
Genotype–phenotype correlations
Additional cautions and considerations
Prenatal diagnosis of mitochondrial disorders
Reporting incidental (secondary) results
Ethical considerations in prenatal testing
References
Chapter 15 Prenatal Diagnosis of Cystic Fibrosis
Genetics and epidemiology
Clinical features
Diagnosis
Treatment
Discovery of the cystic fibrosis gene
The CFTR gene and its protein product
CFTR mutations and variants
Genotype–phenotype correlation
Congenital bilateral absence of the vas deferens
Modifier genes
Ethnic variation in mutation frequencies
Development and implementation of public policy for CF population carrier screening and the core mutation panel
Laboratory methods
Expanded panels
Outcomes of the CF carrier screening program
Special prenatal diagnosis situations
Positive–negative couples
Positive family history
Echogenic bowel
Assisted reproduction and preimplantation diagnosis
Newborn screening
Future directions
References
Chapter 16 Prenatal Diagnosis and the Spectrum of Involvement from Fragile X Mutations
Introduction
Epidemiology
Clinical involvement in those with the full mutation
Clinical phenotype in the premutation
Pathogenesis of the premutation‐associated disorder FXTAS
Neuropathology
Molecular pathogenesis
Molecular prenatal diagnosis methodology
Preimplantation genetic testing and polar body analysis
Neurobiologic advances and targeted treatment in the full mutation
Genetic counseling
Acknowledgments
References
Chapter 17 Prenatal Diagnosis of Fetal Malformations by Ultrasound
Introduction
Craniospinal defects
Neural tube defects
Acrania–exencephaly–anencephaly sequence
Open spina bifida
Encephalocele
Ventriculomegaly – hydrocephaly
Hydranencephaly
Holoprosencephaly
Microcephaly
Corpus callosum agenesis
Posterior fossa malformations
Mega cisterna magna
Blake's pouch cyst
Vermian hypoplasia
Dandy–Walker malformation
Fetal face
Cleft lip and/or palate
Micrognathia and retrognathia
Pulmonary and thoracic abnormalities
Congenital pulmonary airway malformation
Congenital diaphragmatic hernia
Pleural effusions
Congenital high airway obstruction syndrome
Fetal hydrops (immune and nonimmune)
Cardiovascular defects
Cardiac anomalies
Ventricular septal defect
Atrioventricular septal defect
Transposition of the great arteries
Tetralogy of Fallot
Abdominal wall defects
Omphalocele (exomphalos)
Gastroschisis
Body stalk anomaly
Bladder exstrophy and cloacal exstrophy
Gastrointestinal anomalies
Esophageal atresia
Duodenal atresia
Small bowel obstruction
Meconium peritonitis
Abdominal cysts
Kidneys and urinary tract anomalies
Renal development and CAKUT
Dilatation of the renal pelvis
Structural kidney malformations
Renal agenesis
Abnormalities of pelvic migration
Skeletal anomalies
Nuchal translucency
Phenotypic expression in chromosome anomalies
Trisomy 21 – Down syndrome
Trisomy 18 – Edwards syndrome
Trisomy 13 – Patau syndrome
Triploidy
Turner syndrome
References
Chapter 18 Prenatal Diagnosis and Management of Abnormal Fetal Development in the Third Trimester of Pregnancy
Cardiac anomalies
Value of the four‐chamber view in screening for congenital heart disease
Abnormalities in four‐chamber view screening
Hypoplastic left heart
Functional assessment of the fetal heart
Echogenic lung lesions
Bronchopulmonary sequestration
Congenital cystic adenomatoid malformation
Congenital high airway obstruction sequence
Hydrothorax
Congenital diaphragmatic hernia
Anomalies of gastrointestinal tract and abdominal wall
Gastrointestinal obstruction
Abdominal wall defects
Urinary tract anomalies
Incidence and etiology
Diagnosis
Fetal treatment
Prognosis
Central nervous system malformations
Fetal ventriculomegaly
Spina bifida
Absence of cavum septum pellucidum and abnormalities in the midline
Holoprosencephaly
Agenesis of the corpus callosum
Schizencephaly
Absent cavum septum pellucidum
Abnormalities of the posterior cranial fossa and cerebellar anomalies
Parental counseling after diagnosis of fetal brain abnormalities
References
Chapter 19 Prenatal Diagnosis by Fetal Magnetic Resonance Imaging
Introduction
MRI of the fetal central nervous system
Technical issues
Fetal brain MRI: when and why?
Developing brain
Developmental abnormalities
CNS malformations
Ventriculomegaly and genetic disorders
Inborn errors of metabolism
Subependymal cysts
Brain injury
MRI of non‐CNS fetal systems
Technical issues
Fetal neck
Fetal chest
Fetal abdomen and pelvis
Urinary tract pathologies, kidney diseases, and genital malformations
Skeletal malformations
Conclusion
References
Chapter 20 Prenatal Diagnosis of Skeletal Dysplasias and Connective Tissue Disorders
Prenatal sonographic diagnosis of skeletal dysplasias
Abnormal fetal morphology as an unexpected finding
Molecular testing during pregnancy
Estimating the probability of recurrence
Achondroplasia, thanatophoric dysplasia, and hypochondroplasia (FGFR3 disorders)
Prenatal diagnosis
Osteogenesis imperfecta
Prenatal diagnosis
Disorders due to defects in type II collagen (achondrogenesis type 2, hypochondrogenesis, and spondyloepiphyseal dysplasia congenita)
Prenatal diagnosis
Disorders due to defects in the diastrophic dysplasia sulfate transporter gene (achondrogenesis 1B, atelosteogenesis type 2, and diastrophic dysplasia)
Prenatal diagnosis
Joint dislocations: Larsen syndrome and connective tissue disorders
Prenatal diagnosis
Marfan syndrome and Marfan overlap disorders
Pregnancy‐related aspects and prenatal diagnosis
Acknowledgments
References
Chapter 21 Prenatal Diagnosis of Disorders of Carbohydrate Metabolism
Introduction
Glycogen storage diseases
Type I GSD (glucose‐6‐phosphatase and glucose‐6‐phosphate translocase deficiency, von Gierke disease)
Type II GSD (acid ?‐glucosidase deficiency, Pompe disease)
Type III GSD (debrancher deficiency, limit dextrinosis, Cori or Forbes disease)
Type IV GSD (branching enzyme deficiency, amylopectinosis, or Andersen disease)
Type V GSD (muscle phosphorylase deficiency, McArdle disease, myophosphorylase deficiency)
Type VI GSD (liver phosphorylase, Hers disease)
Type VII GSD (phosphofructokinase deficiency, Tarui disease)
Type IX GSD (phosphorylase b kinase deficiency)
Glycogen synthase deficiency
Hepatic glycogenosis with renal Fanconi–Bickel syndrome
Disorders of galactose metabolism
Galactosemia with transferase deficiency
Galactokinase deficiency
Uridine diphosphate galactose‐4‐epimerase (UDPgal‐4‐epimerase) deficiency
Disorders of fructose metabolism
Essential fructosuria
Hereditary fructose intolerance (fructose‐1‐phosphate aldolase B deficiency)
Disorders of gluconeogenesis
Fructose‐1,6‐bisphosphatase deficiency
Phosphoenolpyruvate carboxykinase deficiency
Pentosuria
Acknowledgments
References
Chapter 22 Disorders of Metabolism of Amino Acids and Related Compounds
Introduction
Inborn errors of metabolism
Amino acid disorders
Methods of prenatal screening of amino acid disorders
The placenta as a filter
Maternal nutrition and amino acid disorders: potential impact on the fetus
Intoxication disorders
Urea cycle disorders
Other disorders presenting with hyperammonemia or involving urea cycle intermediates
Disorders of ornithine metabolism
Sulfite oxidase deficiency
Nonketotic hyperglycinemia
Mevalonic aciduria
4‐Hydroxybutyric aciduria (succinic semialdehyde dehydrogenase deficiency)
Disorders of organic acids
Other disorders of catabolism of branched‐chain amino acids
Disorders of sulfur amino acid metabolism
Disorders of energy production
l‐2‐Hydroxyglutaric aciduria
d‐2‐Hydroxyglutaric aciduria
Glutaric aciduria type II (multiple acyl‐CoA dehydrogenase disorder)
Very rare amino acid disorders
Hypervalinemia (a disorder of valine metabolism)
Hypermethioninemia due to methionine adenosyltransferase deficiency
Combined d‐2‐ and l‐2‐hydroxyglutaric aciduria
Methylenetetrahydrofolate reductase deficiency
Prolidase deficiency
Disorders of proline metabolism
Disorders of renal amino acid transport
In conclusion
References
Chapter 23 The Mucopolysaccharidoses: Prenatal Diagnosis, Neonatal Screening and Emerging Therapies
Introduction
Disease and biochemical characteristics
Clinical and biochemical fundamentals
Structure and function of glycosaminoglycans
Prenatal diagnosis
Clinical characteristics and disease pathogenesis
Clinical heterogeneity
Genetic heterogeneity
Genotype–phenotype correlations
Mucopolysaccharidose disease pathogenesis
Pathophysiology of disease
Pathogenesis of MPS skeletal disease
Pathogenesis of MPS nervous system disease
Postnatal MPS therapeutics
Newborn screening
Fetal considerations
Future directions
References
Chapter 24 Prenatal Diagnosis of the Peroxisomal and Mitochondrial Fatty Acid Oxidation Deficiencies
Introduction
Mitochondrial versus peroxisomal fatty acid β‐oxidation
Mitochondrial fatty acid β‐oxidation disorders
Primary carnitine deficiency (OCTN2 deficiency) (OMIM 212140)
Carnitine palmitoyl transferase‐1 A deficiency (OMIM 600528)
Carnitine–acylcarnitine translocase deficiency (OMIM 212138)
Carnitine palmitoyl transferase 2 deficiency (OMIM 600649, 600650, 255110, 608836)
Very long‐chain acyl‐CoA dehydrogenase deficiency (OMIM 201475)
Medium‐chain acyl‐CoA dehydrogenase deficiency (OMIM 201450)
Mitochondrial trifunctional protein deficiency (OMIM 600890)
Secondary disorders of mitochondrial fatty acid oxidation
Peroxisomal fatty acid β‐oxidation disorders
Primary peroxisomal fatty acid oxidation disorders
X‐linked adrenoleukodystrophy (OMIM 300100)
Acyl‐CoA oxidase 1 deficiency (OMIM 264470)
Acyl‐CoA oxidase 2 deficiency (OMIM 601641)
D‐bifunctional protein deficiency (OMIM 261515)
Sterol carrier protein X deficiency (OMIM 613724)
2‐Methylacyl‐CoA racemase deficiency (OMIM 604489)
PMP70 deficiency (OMIM 170995) and ACBD5 deficiency (OMIM 616618)
Secondary disorders of peroxisomal fatty acid oxidation
References
Chapter 25 Prenatal Diagnosis of Disorders of Lipid Metabolism
Introduction
Lipoprotein‐associated disorders
Defects in the metabolism of glycosphingolipids
Structure of glycosphingolipids
Function and distribution of glycosphingolipids
Biosynthesis of glycosphingolipids
Defects in the biosynthesis of glycosphingolipids
The lysosomal catabolism of glycosphingolipids
GM1‐gangliosidosis/mucopolysaccharidosis IVB (Morquio B)
GM2‐gangliosidoses
Tay–Sachs disease: mutations in HEXA gene (?‐subunit) (OMIM: 272800)
B1 variant
Pseudodeficiency
Hexosaminidase S
Sandhoff disease: mutations in HEXB (β‐subunit) (GM2‐gangliosidosis 0 variant) (OMIM: 268800)
Variant AB
Fabry disease
Gaucher disease
Metachromatic leukodystrophy
Multiple sulfatase deficiency
Krabbe disease (globoid cell leukodystrophy)
Niemann–Pick disease
Niemann–Pick disease types A and B (acid sphingomyelinase deficiency)
Niemann–Pick type C
Farber disease
Lysosomal acid lipase deficiency: Wolman disease and cholesteryl ester storage disease
The neuronal ceroid lipofuscinoses
Acknowledgments
References
Chapter 26 Prenatal Diagnosis of Primary Immunodeficiency Diseases
Family history
Specific immune defects
Lymphocyte deficiencies
T‐cell and combined deficiencies
Antibody deficiencies
Phagocyte deficiencies
Defects with autoimmunity or immune dysregulation
Complement deficiencies
Additional syndromic immune defects
References
Chapter 27 Prenatal Diagnosis of the Hemoglobinopathies
Introduction
Clinical types
?‐Thalassemia
Hb Bart's hydrops fetalis syndrome
Hb H disease
β‐Thalassemia
β‐Thalassemia major
β‐Thalassemia intermedia
Hb E disorders
Hb E/β‐thalassemia
Hb AE Bart's disease
Hb EF Bart's disease
Hb E/E plus ?CS?/?CS?
Sickle cell disorders
Sickle cell anemia
Hb S/β‐thalassemia
Hb S/fδβ‐thalassemia
Hb S/Hb C
Hb S/Hb E
Hb S/Hb D‐Punjab
Hb S/Hb O‐Arab
Hb S/Hb C‐Harlem
Hb S/Hb S‐Southend
Hb S‐Antilles
Hb S‐Oman
Other sickling variants
Hb S/other rare β‐chain variants
Carrier screening
Reduced red cell indices with a raised Hb A2 value
Reduced red cell indices with a normal Hb A2 value
Strategy for fetal diagnosis
Approaches to prenatal diagnosis
Amniotic fluid DNA
Chorionic villus DNA
Noninvasive prenatal diagnosis
Preimplantation diagnosis
DNA diagnosis of the hemoglobinopathies
?‐Thalassemia
β‐Thalassemia
The δβ‐thalassemias, Hb Lepore, and hereditary persistence of fetal hemoglobin disorders
Abnormal hemoglobins
Hb S
Hb C
Hb D‐Punjab and Hb O‐Arab
Hb E
Diagnostic pitfalls and best practice for fetal diagnosis
Maternal DNA contamination
Technical errors
Diagnostic error rate
Guidelines for best practice
Summary
References
Chapter 28 Prenatal Diagnosis of Inherited Disorders of Folate and Cobalamin Metabolism
Inborn errors of folate metabolism
Hereditary malabsorption of folate
Cerebral folate deficiency
Glutamate formiminotransferase deficiency
Methylenetetrahydrofolate reductase deficiency
Dihydrofolate reductase deficiency
MTHFD1 deficiency
Methenyltetrahydrofolate synthetase deficiency
Inborn errors of cobalamin metabolism
Disorders of cobalamin uptake
Transcobalamin deficiency
Transcobalamin receptor deficiency
Disorders of cobalamin utilization
Isolated methylmalonic aciduria
Isolated methylcobalamin deficiency
Combined methylmalonic aciduria and homocystinuria
Prenatal diagnosis and fetal therapy
References
Chapter 29 Fetal Surgery
Introduction
Brief history of fetal surgery
Ethical considerations
Imaging principles for fetal intervention and surgical procedures
Control of fetal pain
Closed fetal therapies
Conditions treated using fetoscopic procedures
Placental laser photocoagulation for twin‐to‐twin transfusion syndrome
Placental laser photocoagulation for twin anemia polycythemia sequence
Placental laser photocoagulation in monochorionic diamniotic twin pregnancies with selective fetal growth restriction
Congenital diaphragmatic hernia
Tracheal occlusion in human fetuses
Urinary tract obstruction
Fetal pleural effusion
Congenital cystic adenomatoid malformation
Other conditions treated with fetoscopic procedures
Amniotic band syndrome
Vasa previa
Chorioangioma
Selective termination in monochorionic gestation
Open fetal surgery technique and complications
Conditions treated with open fetal surgery
Myelomeningocele
Sacrococcygeal teratoma
Other potentially beneficial fetal interventions
Congenital heart defect interventions
Ex utero intrapartum treatment
Recent advances in fetal surgery
Complex fetoscopic surgery
References
Chapter 30 In Utero Stem Cell Transplantation, Enzyme Replacement, and Gene Therapy
Introduction to in utero therapy
In utero hematopoietic stem cell transplantation
Background and preclinical studies
Preclinical studies in large‐animal models
Clinical studies
Future of in utero hematopoietic stem cell transplantation
In utero enzyme replacement therapy
Background
Preclinical studies
Clinical studies
Future of in utero enzyme replacement therapy
In utero gene therapy
Background
Risks of in utero gene therapy
Preclinical studies of in utero gene therapy
Clinical studies of in utero gene therapy
Conclusions
References
Chapter 31 Maternal Genetic Disorders That Affect Fetal Health
Introduction to inherited metabolic disorders
A genetic disorder with a teratogenic effect on the fetus: phenylketonuria
Genetic disorders precipitated by catabolic states including the late third trimester, intrapartum, and the puerperium: disabled protein breakdown
Urea cycle disorders
Other disorders of amino acid metabolism
Organic acid disorders of protein metabolism
Disorders of energy metabolism aggravated by maternal–fetal anabolic states
Disorders of fatty acid and lipid metabolism
Sphingolipidoses
Disorders of carbohydrate metabolism
Disorders of metal metabolism
Wilson disease
Connective tissue disorders
Ehlers–Danlos syndrome
Marfan syndrome
Loeys–Dietz syndrome
Maternal skeletal dyplasias (chondrodystrophies)
Turner syndrome (monosomy X)
Cystic fibrosis
Neuromuscular disorders
Myotonic dystrophy
Duchenne/Becker muscular dystrophy
Hematologic disorders
Sickle cell disorders
Fanconi anemia
Hereditary coagulopathies and inherited platelet disorders
References
Chapter 32 Pregnancy Termination for Genetic Disorders: Indications and Complications
Introduction
First‐trimester pregnancy termination techniques
Suction aspiration
Medical abortion
Second‐trimester techniques
Dilation and evacuation
Systemic abortifacients
Intra‐amniotic abortifacients and hysterotomy/hysterectomy
Selective abortion/fetal reduction in multiple gestations
First‐trimester selective fetal reduction
Second‐trimester selective fetal reduction
Counseling patients about multifetal pregnancy selective reduction procedures
Conclusions
Acknowledgments
References
Chapter 33 Providing Supportive Psychosocial Care to Parents after Perinatal Loss
Introduction
Perinatal Loss
Complex grief after perinatal loss
Highly intense grief and associated health issues
Termination of pregnancy for severe or lethal fetal anomaly
Needs of healthcare providers
Hutti Theoretical Framework of Perinatal Grief Intensity
Reality
Congruence
Confront others
Use of the Hutti Theoretical Framework in clinical practice
Perinatal Grief Intensity Scale
Communicating bad news to parents
Preparing parents for the experience of perinatal loss
Labor and birth
After hospital discharge
Subsequent pregnancy
Interventions for high‐quality perinatal bereavement care
Regoaling
Conclusion
References
Chapter 34 Prenatal Diagnosis of Fetal Infection
Prenatal diagnosis of fetal toxoplasmosis
Parasitology
Epidemiology
Risk factors
Prenatal diagnosis
In utero therapeutic options
Management at birth
Prevention of fetal toxoplasmosis: education
Summary
Prenatal diagnosis of fetal cytomegalovirus infection
Virology
Epidemiology
Pathogenesis of congenital infection
Pathology
Congenital infection
Clinical maternal manifestations
Serology
Management
Diagnosis of congenital CMV infection in the fetus
Forming a prognosis for fetal CMV infection
Treatment of congenital CMV infection
Prevention of CMV infection
Screening for congenital CMV infection
Prenatal diagnosis of congenital rubella
Risk of fetal infection
Definition of maternal infection
Prenatal diagnosis of fetal infection
Summary
Prenatal diagnosis of fetal varicella infection
Virologic bases
Epidemiology
Clinical aspects of maternal infection
Consequences for the pregnancy and the fetus
Pathogenesis of fetal VZV infection
Diagnosis of fetal VZV infection
Frequency of VZV transmission
Treatment and prevention of VZV
Summary
Prenatal diagnosis of human parvovirus B19 infection
Epidemiology, maternal infection, and vertical transmission of parvovirus B19
Prenatal diagnosis of parvovirus B19
Treatment of parvovirus B19
Prenatal diagnosis of Zika virus
Maternal infection with Zika virus
Infection of the fetus with the Zika virus
Prenatal counseling in the midst of the SARS‐CoV‐2 pandemic
Acknowledgments
References
Chapter 35 Medicolegal Aspects of Prenatal Diagnosis
General concepts of medical malpractice
The constitutional right of privacy in reproductive decisions
The role of informed consent in medical treatment
Suits for wrongful birth and wrongful life
Wrongful pregnancy – the parents' claim, usually after having a healthy, but unwanted child
Wrongful birth – the parents' claim after having a child with a genetic disorder or congenital anomaly
Wrongful life – the child's claim
Wrongful life claims against the parents
The impact of concerns about abortion on wrongful birth and wrongful life claims
Future trends
Summary of alleged negligent acts and outcomes in wrongful birth and wrongful life suits
Statutes addressing claims for wrongful birth and wrongful life as well as abortion in the case of fetal anomaly
References
Chapter 36 Prenatal and Preimplantation Diagnosis: International Policy Perspectives
Introduction
Prenatal diagnosis
Legal approaches
Professional guidelines
Preimplantation genetic diagnosis
Legal approach
Professional guidelines
Hybrid regulatory approach
Sex selection in prenatal diagnosis and preimplantation genetic diagnosis
Emerging technologies, new issues?
References
Chapter 37 Ethical Issues in the Diagnosis and Management of Genetic Disorders in the Fetus
Professional ethics in obstetrics
The ethical principle of beneficence
The ethical principle of respect for autonomy
The ethical concept of the fetus as a patient
Clinical applications of professional ethics in obstetrics
Prenatal genetic counseling
Diagnosis of genetic disorders in the fetus
Preimplantation diagnosis and the “savior sibling”
Management of pregnancies complicated by genetic disorders
Ethical issues in clinical innovation and research
Genomic alteration research
Maternal–fetal medical and surgical intervention for fetal benefit innovation and research
Human embryonic stem cell research
Conclusion
References
Index
EULA

Citation preview

Genetic Disorders and the Fetus

Dedicated to

Laura and Francia For their love, support, and understanding

“Make assurance double sure.” Shakespeare, Macbeth

Genetic Disorders and the Fetus Diagnosis, Prevention, and Treatment EIGHTH EDITION

EDITED BY

Aubrey Milunsky, MB BCh, DSc, FRCP, FACMG, DCH Professor of Obstetrics and Gynecology Tufts University School of Medicine Formerly Professor of Human Genetics, Pediatrics, Obstetrics and Gynecology Boston University School of Medicine Founder and Co-Director, Center for Human Genetics Cambridge, MA, USA

Jeff M. Milunsky, MD, FACMG Professor of Obstetrics and Gynecology Tufts University School of Medicine Formerly Professor of Genetics and Genomics, and Pediatrics Boston University School of Medicine Co-Director, Center for Human Genetics Director, Clinical and Molecular Genetics Cambridge, MA, USA

This eighth edition first published 2021 © 2021 Aubrey Milunsky and Jeff M. Milunsky Edition History Previous editions: © 2016, 2010 Aubrey Milunsky and Jeff M. Milunsky; © 2004, 1998, 1992, 1986, 1979 Aubrey Milunsky All rights reserved. No part of this publication may be reproduced, stored in a retrieval system, or transmitted, in any form or by any means, electronic, mechanical, photocopying, recording or otherwise, except as permitted by law. Advice on how to obtain permission to reuse material from this title is available at http://www.wiley.com/go/permissions. The right of Aubrey Milunsky and Jeff M. Milunsky to be identified as the authors of the editorial material in this work has been asserted in accordance with law. Registered Offices John Wiley & Sons, Inc., 111 River Street, Hoboken, NJ 07030, USA John Wiley & Sons Ltd, The Atrium, Southern Gate, Chichester, West Sussex, PO19 8SQ, UK Editorial Office 9600 Garsington Road, Oxford, OX4 2DQ, UK For details of our global editorial offices, customer services, and more information about Wiley products visit us at www.wiley.com. Wiley also publishes its books in a variety of electronic formats and by print-on-demand. Some content that appears in standard print versions of this book may not be available in other formats. Limit of Liability/Disclaimer of Warranty The contents of this work are intended to further general scientific research, understanding, and discussion only and are not intended and should not be relied upon as recommending or promoting scientific method, diagnosis, or treatment by physicians for any particular patient. In view of ongoing research, equipment modifications, changes in governmental regulations, and the constant flow of information relating to the use of medicines, equipment, and devices, the reader is urged to review and evaluate the information provided in the package insert or instructions for each medicine, equipment, or device for, among other things, any changes in the instructions or indication of usage and for added warnings and precautions. While the publisher and authors have used their best efforts in preparing this work, they make no representations or warranties with respect to the accuracy or completeness of the contents of this work and specifically disclaim all warranties, including without limitation any implied warranties of merchantability or fitness for a particular purpose. No warranty may be created or extended by sales representatives, written sales materials or promotional statements for this work. The fact that an organization, website, or product is referred to in this work as a citation and/or potential source of further information does not mean that the publisher and authors endorse the information or services the organization, website, or product may provide or recommendations it may make. This work is sold with the understanding that the publisher is not engaged in rendering professional services. The advice and strategies contained herein may not be suitable for your situation. You should consult with a specialist where appropriate. Further, readers should be aware that websites listed in this work may have changed or disappeared between when this work was written and when it is read. Neither the publisher nor authors shall be liable for any loss of profit or any other commercial damages, including but not limited to special, incidental, consequential, or other damages. Library of Congress Cataloging-in-Publication Data Names: Milunsky, Aubrey, editor. | Milunsky, Jeff M., editor. Title: Genetic disorders and the fetus : diagnosis, prevention, and treatment / edited by Aubrey Milunsky, MB BCh, DSc, FRCP, FACMG, DCH and Jeff M. Milunsky, MD, FACMG. Description: Eighth edition. | Hoboken, NJ : Wiley-Blackwell, 2021. Identifiers: LCCN 2021058739 (print) | LCCN 2021058740 (ebook) | ISBN 9781119676935 (hardback) | ISBN 9781119676973 (adobe pdf) | ISBN 9781119676959 (epub) Subjects: LCSH: Prenatal diagnosis. | Fetus–Diseases–Genetic aspects. | Fetus–Abnormalities–Genetic aspects. Classification: LCC RG628 .G46 2021 (print) | LCC RG628 (ebook) | DDC 618.3/2075–dc23 LC record available at https://lccn.loc.gov/2021058739 LC ebook record available at https://lccn.loc.gov/2021058740 Cover Design: Wiley Cover Image: © (Left column): Rasi Bhadramani/iStock/Getty Images. (Top row)(left) Roland Axt-Fliedner and Aline Wolter; (center) Liesbeth van Leeuwen, Malou A. Lugthart, and Eva Pajkrt; (right) Liesbeth van Leeuwen, Malou A. Lugthart, and Eva Pajkrt. (Middle row)(left) Nadine Girard and Kathia Chaumoitre; (center) Nadine Girard and Kathia Chaumoitre; (right) Michael A. Belfort and Alireza A. Shamshirsaz. (Bottom row)(left) Anver Kuliev and Svetlana Rechitsky. Set in 9.5/12pt MinionPro by SPi Global, Chennai, India

10 9 8 7 6 5 4 3 2 1

Contents

Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xiii Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xv 1 Genetic Counseling: Preconception, Prenatal, and Perinatal . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1 Aubrey Milunsky and Jeff M. Milunsky 2 Preimplantation Genetic Testing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 102 Anver Kuliev and Svetlana Rechitsky 3 Amniotic Fluid Constituents, Cell Culture, and Neural Tube Defects . . . . . . . . . . . . . . . . . . . . . . . . . . 140 Daniel L. Van Dyke and Aubrey Milunsky 4 Molecular Aspects of Placental Development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 197 Wendy P. Robinson and Deborah E. McFadden 5 Fetal Origins of Adult Health and Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 214 Michael G. Ross and Mina Desai 6 Maternal Serum Screening for Chromosomal Abnormalities and Neural Tube Defects . . . . . . . . . . 240 Howard Cuckle 7 Noninvasive Screening for Aneuploidy Using Cell-Free Placental DNA . . . . . . . . . . . . . . . . . . . . . . . . 301 Lorraine Dugoff 8 Noninvasive Prenatal Diagnosis and Screening for Monogenic Disorders Using Cell-Free DNA . . 318 Ignatia B. Van den Veyver, Natalie Chandler and Lyn S. Chitty 9 Amniocentesis, Chorionic Villus Sampling, and Fetal Blood Sampling . . . . . . . . . . . . . . . . . . . . . . . . . 346 Anthony O. Odibo 10 Prenatal Diagnosis of Neural Tube Defects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 375 Aubrey Milunsky 11 Prenatal Diagnosis of Chromosomal Abnormalities through Chorionic Villus Sampling and Amniocentesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 404 Peter A. Benn 12 Prenatal Diagnosis of Sex Chromosome Abnormalities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 499 Jeff M. Milunsky 13 Prenatal Diagnosis of Chromosomal Abnormalities: From Karyotype to Microarray . . . . . . . . . . . . 547 Brynn Levy

v

vi

Contents

14 Molecular Genetics and Prenatal Diagnosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 572 Jeff M. Milunsky 15 Prenatal Diagnosis of Cystic Fibrosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 611 Wayne W. Grody 16 Prenatal Diagnosis and the Spectrum of Involvement from Fragile X Mutations . . . . . . . . . . . . . . . . 629 Randi J. Hagerman and Paul J. Hagerman 17 Prenatal Diagnosis of Fetal Malformations by Ultrasound . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 647 Liesbeth van Leeuwen, Malou A. Lugthart and Eva Pajkrt 18 Prenatal Diagnosis and Management of Abnormal Fetal Development in the Third Trimester of Pregnancy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 701 Roland Axt-Fliedner and Aline Wolter 19 Prenatal Diagnosis by Fetal Magnetic Resonance Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 760 Nadine Girard and Kathia Chaumoitre 20 Prenatal Diagnosis of Skeletal Dysplasias and Connective Tissue Disorders . . . . . . . . . . . . . . . . . . . . 783 Andrea Superti-Furga and Sheila Unger 21 Prenatal Diagnosis of Disorders of Carbohydrate Metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 803 Deeksha Sarihyan Bali, Stephanie L. Austin and Priya S. Kishnani 22 Disorders of Metabolism of Amino Acids and Related Compounds . . . . . . . . . . . . . . . . . . . . . . . . . . . 831 Andrea Gropman, Georgianne Arnold and Jerry Vockley 23 The Mucopolysaccharidoses: Prenatal Diagnosis, Neonatal Screening and Emerging Therapies . . . 868 Lorne A. Clarke 24 Prenatal Diagnosis of the Peroxisomal and Mitochondrial Fatty Acid Oxidation Deficiencies . . . . 890 Ronald J.A. Wanders and Hans R. Waterham 25 Prenatal Diagnosis of Disorders of Lipid Metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 910 James E. Davison, Steven E. Humphries, Bryan G. Winchester and Sara E. Mole 26 Prenatal Diagnosis of Primary Immunodeficiency Diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 982 Jennifer M. Puck 27 Prenatal Diagnosis of the Hemoglobinopathies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1002 John M. Old and Jan Traeger-Synodinos 28 Prenatal Diagnosis of Inherited Disorders of Folate and Cobalamin Metabolism . . . . . . . . . . . . . . . 1035 David S. Rosenblatt and David Watkins 29 Fetal Surgery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1050 Michael A. Belfort and Alireza A. Shamshirsaz 30 In Utero Stem Cell Transplantation, Enzyme Replacement, and Gene Therapy . . . . . . . . . . . . . . . . . 1105 Tippi C. MacKenzie and Marisa E. Schwab 31 Maternal Genetic Disorders That Affect Fetal Health . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1120 Karin J. Blakemore 32 Pregnancy Termination for Genetic Disorders: Indications and Complications . . . . . . . . . . . . . . . . 1158 Lee P. Shulman

Contents vii

33 Providing Supportive Psychosocial Care to Parents after Perinatal Loss . . . . . . . . . . . . . . . . . . . . . . . 1179 Marianne H. Hutti 34 Prenatal Diagnosis of Fetal Infection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1197 Marianne Leruez-Ville, Valentine Faure-Bardon and Yves G. Ville 35 Medicolegal Aspects of Prenatal Diagnosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1236 Ellen Wright Clayton 36 Prenatal and Preimplantation Diagnosis: International Policy Perspectives . . . . . . . . . . . . . . . . . . . . 1250 Minh Thu Minh Nguyen and Bartha Maria Knoppers 37 Ethical Issues in the Diagnosis and Management of Genetic Disorders in the Fetus . . . . . . . . . . . . 1267 Frank A. Chervenak and Laurence B. McCullough Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1294

Preface

Certainty and trust are the hallmarks of prenatal diagnosis where doubt and ambiguity are outlaws. At no time was that more apparent than when AM made his first prenatal genetic diagnosis over 50 years ago. “Are you sure?” were that patient’s first words, intoned with deep anxiety. Those words underscored the still cogent need for accuracy in prenatal diagnosis, where mostly a single report would be final. That was a time when prenatal diagnosis depended on amniocentesis-based study, yielding an accuracy rate exceeding 99 percent. Few laboratory tests, then or now, concerned with profoundly important decision-making equal this enviable accuracy. Variants of uncertain significance had not yet entered the genetic lexicon. Today, however, with the newer, beguiling, noninvasive technology, amniocentesis and chorionic villus sampling (CVS) use has declined dramatically. On the altars of convenience and expediency, women are choosing or simply encouraged to have noninvasive prenatal testing, often blissfully unaware that about half of all chromosomal abnormalities detectable through amniocentesis or CVS and authoritatively discussed, would be missed. There is room for concern given the current consensus that all women should be offered either of these two procedures. Similarly, all women with risks of having offspring with a monogenic disorder and a known pathogenic variant should be informed about and offered the option of preimplantation genetic testing (PGT). The extensive and long-established experience of the authors with PGT enshrine this recommendation. Expanded carrier screening should make this a more frequently addressed option. Couples should, however, be carefully counseled about the limitations of such screening. All the facts, figures, guidelines, and recommendations have been updated in this edition, and will

assist the expanded healthcare team in providing optimal care to all patients. Coupled with remarkable advances in technology, prenatal diagnosis has undergone a revolution. Thus far, and continually increasing, more than 4,331 culprit genes and 6,739 associated phenotypes for an extensive array of genetic disorders have been identified. Consequently, since physicians in all medical specialties encounter genetic disorders for which a molecular diagnosis is available, awareness of the options for prenatal diagnosis or PGT has become especially important. Pari passu with these technical advances has come the opportunity to avoid and prevent the occurrence of many lethal and seriously disabling genetic diseases. This progress means that physicians in all specialties incur responsibility (and inevitable liability) to acquire new knowledge of genetic disorders and offer appropriate tests or refer patients for evaluation and genetic counseling. More than 60 million people worldwide are estimated to have DNA studies in the next 5 years. Expanded carrier testing, maternal cell-free plasma DNA testing, whole-exome sequencing, next-generation sequencing, single-cell molecular diagnosis, and advanced fetal imaging, all fully presented in this volume, now complement the well-established procedure for prenatal diagnosis and PGT. Whole-genome analysis for prenatal diagnosis (even if advisable) simply awaits further technical advances. Chromosomal microarrays and whole-exome sequencing have propelled an even greater prenatal diagnostic reach, especially in the face of fetal structural, including skeletal, abnormalities, given the advances in ultrasound and magnetic resonance imaging, so expertly covered in this volume by authors from three European countries. The fetal anomaly scan has to an important extent superseded amniotic fluid 𝛼-fetoprotein for ix

x Preface

the diagnosis of neural tube defects. Nevertheless, the significant worldwide prevalence of neural tube defects makes maternal serum 𝛼-fetoprotein screening a continuing vital test, critically evaluated here by a pioneer. The diagnosis of neural tube defects and their consequences, as well as the frequently failed efforts at prevention, are thoroughly detailed. Knowledge of the common sex chromosome aneuploidies and some of the rarer variants continues to expand. Up-to-date summaries of all these disorders are presented with specific recommendations for genetic counseling. Molecular prenatal diagnosis has now become routine and the multiple technologies utilized, with their benefits and limitations along with clinically based caveats and considerations, are presented in a significantly updated chapter. The advent of next-generation sequencing has, by targeting panels or whole-exome approaches, resulted in more opportunities for avoidance and prevention through prenatal diagnosis. These widely available technologies not only address previously diagnosed childhood-onset disorders, but also those of adult onset, including cardiomyopathies, malignancies, and neurologic disorders. Progress and refinement in the diagnosis and management of a wide range of metabolic disorders are fully and authoritatively updated, and equally complemented by the detailed in-depth advances in molecular diagnostics that include the hemoglobinopathies, fragile X syndrome, cystic fibrosis, disorders of folate metabolism, and the immunodeficiency diseases. Pregnancy termination is a sad but fortunately uncommon consideration following prenatal diagnostic studies. The techniques and complications are fully discussed and complemented by insightful senior experience with the management of grief after pregnancy and perinatal loss. Discussion about the care and management of mothers with genetic disorders that affect fetal health and those who transmit infection to the fetus is sharply focused on diagnosis, prevention, avoidance, and treatment. Fetal health is doubly important given the known fetal origins of adult disease that go far beyond hypertension, obesity, and diabetes, to include the epigenetic phenomena induced by the maternal pregnancy environment. A thorough

exposition on the importance of placental development, structure, function, genetics, and pathology on fetal growth and development is expertly presented in this edition. Steadily, but surely, fetal gene therapy via hematopoietic stem cell transplantation is taking root, while a remarkable chapter on fetal surgery by a leader in the field points to new brave surgical remedies. While all authors acknowledge continuing progress in molecular genetics, inconclusive results due to variants of uncertain significance are not infrequent. Laboratory conclusions can be further compounded by a host of issues that potentially befuddle interpretation. Commonly encountered issues include delineation of normal variation or polymorphisms, difficulty determining the pathogenicity of variants, depth of sequencing coverage, regions of high GC content, mosaicism, DNA contamination, digenic inheritance, locus heterogeneity, and false-positive and false-negative results. The concurrence of an incidental (secondary) finding on fetal DNA analysis will predictably arouse great parental anxiety. Current laws and public policy regarding prenatal diagnosis and PGT in 16 countries are examined in detail regarding international differences, with special reference to the guidelines relevant to the emerging technologies. A senior physician-lawyer, in reviewing the important principles in the torts of wrongful birth and wrongful life, focused on the potential liability of those involved in reproductive medicine. Professional ethics in obstetrics, with emphasis on the ethical principles of beneficence and autonomy and the ethical concept of the fetus as a patient, receive in-depth discussion by doyens in this field. This volume is a major repository of facts about prenatal diagnosis and provides a critical analysis and synthesis of established and new knowledge based on the long experience of the senior contributing authorities in their respective fields. In addition, a broad international perspective is presented with contributions from recognized international experts from nine countries. The guidance provided and the insights and perspectives of these authors make this volume a valuable and indispensable resource for all those whose focus is securing fetal health through prenatal diagnosis or PGT.

Preface xi

This text is very well referenced and replete with evidence-based guidance and reflective of the lifetime experience and wisdom of distinguished senior authors. This edition encompasses 152 tables, 167 figures, and over 10,000 references. A valuable index will facilitate the reader’s search for specific information. The major technologic advances in genetics have made the requirement for preconception, prenatal, and perinatal genetic counseling of paramount importance. Even though the underlying principles

and prerequisites are well established, the many advances have introduced a panoply of new challenges discussed in detail in a comprehensive, heavily referenced opening chapter. We are in the golden era of human genetics, and through new discoveries and insights have increased opportunities for the diagnosis, prevention, and treatment of many serious and lethal genetic disorders. Aubrey Milunsky and Jeff M. Milunsky Cambridge

Acknowledgments

This eighth edition marks the 42nd year of this text and reflects the continuing remarkable advances made in achieving accurate prenatal and preimplantation diagnoses. The first book on this subject (The Prenatal Diagnosis of Hereditary Disease) was published some 48 years ago (by AM). The distillation of accrued biological, technological, ethical, and legal knowledge has graced these pages and enriched the reference value of these editions. The wisdom, insight, perspective, expertise, experience, and knowledge of these senior contributing authors has made these volumes a valuable and authoritative text. Moreover, these authors have again provided an international perspective, this edition having contributions from nine countries. Expert care in human genetics, maternal–fetal medicine, and perinatal medicine has demanded knowledge, up-to-date information, guidance, and expertise in each volume. This has been achieved only by the willingness of the internationally recognized authoritative authors who have taken the time to share their knowledge, experience, and wisdom. For this we are most appreciative. We are also grateful and indebted to our friends and colleagues who have died and who were

expert contributing authors to earlier editions. We remember them with pride and sadness: Bruno Brambati, MD, David J.H. Brock, PhD, Jacob A. Canick, PhD, Louis Dalliaire, MD, PhD, Sherman Elias, MD, H.J. Evans, PhD, FRSE, John C. Fletcher, PhD, Fredric Frigoletto, MD, Albert B. Gerbie, MD, Leonard A. Herzenberg, PhD, George Hug, MD, Lillian Y.F. Hsu, MD, Mary Z. Pelias, PhD, JD, Arthur Robinson, MD, Richard H. Schwarz, MD, Margery W. Shaw, MD, JD, Irving Umansky, MD, Yury Verlinsky, PhD, and Dorothy C. Wertz, PhD. It is possible that we are unaware of the passing of a few authors and regret any omission. We remain eternally grateful to all these remarkable physicians and scientists. AM deeply appreciates the exemplary work and dedication to perfection of his assistant Marina Nguyen. JM is very grateful for the precision, accuracy, and patience of his assistant Emma Rebholz during the writing and editing of this text.

Aubrey Milunsky Jeff M. Milunsky

xiii

Contributors

Georgianne L. Arnold,

MD Professor of Pediatrics University of Pittsburgh School of Medicine Clinical Director, Division of Medical Genetics Children’s Hospital Pittsburgh Pittsburgh, PA, USA

Stephanie L. Austin,

MS, MA, CGC

Genetic Counsellor Senior Research Program Leader Division of Medical Genetics Duke University Durham, NC, USA

Roland M. Axt-Fliedner,

MD, PhD Professor of Obstetrics and Gynecology Justus-Liebig-University Head Division of Prenatal Medicine and Fetal Therapy University Hospital Giessen, Germany

Deeksha Sarihyan Bali,

PhD, FACMGG Professor of Pediatrics Laboratory Director Biochemical Genetics Laboratory Division of Medical Genetics Duke Health Durham, NC, USA

Michael A. Belfort, MBBCH, D. Mid. COG(SA), D.A (SA), MD, PhD, FRCOG, FRCSC, FACOG Professor and Ernst Bertner Chair, Department of Obstetrics and Gynecology Secondary appointments: Departments of Anesthesiology, Surgery, Neurosurgery Baylor College of Medicine Obstetrician and Gynecologist-in-Chief FB McGuyer and Family Chair in Fetal Surgery Medical Director, Texas Children’s Fetal Center

Texas Children’s Hospital Houston, TX, USA

Peter A. Benn, BSc, MSc, PhD, FACMG, DSc Professor Emeritus Department of Genetics and Genome Sciences University of Connecticut Health Center Farmington, CT, USA Karin J. Blakemore,

MD Professor of Gynecology and Obstetrics, Oncology, and Medical Genetics Director, Prenatal Genetics Center Division of Maternal-Fetal Medicine Department of Gynecology and Obstetrics Johns Hopkins University School of Medicine Baltimore, MD, USA

Natalie Chandler, FRCPath Senior Clinical Scientist North Thames Genomic Laboratory Hub Great Ormond Street Hospital for Children NHS Foundation Trust London, UK Kathia Chaumoitre,

MD, PhD

Professor of Paediatric Radiology Imaging Department North Hospital Aix-Marseille University Marseille, France

Frank A. Chervenak, MD, MMM Professor and Chair Department of Obstetrics and Gynecology Associate Dean, International Medicine Zucker School of Medicine at Hofstra/Northwell Lenox Hill Hospital New York, NY, USA

xv

xvi

Contributors

Lyn S. Chitty, PhD, MB, BS, MRCOG Professor of Genetics and Fetal Medicine Medical Director North Thames Genomic Laboratory Hub Great Ormond Street Hospital for Children Genetics and Genomic Medicine, UCL Institute of Child Health London, UK Lorne A. Clarke,

MDCM, FRCPC, FCCMG Professor of Medical Genetics University of British Columbia Department of Medical Genetics British Columbia Children’s Hospital Research Institute Vancouver, British Columbia, Canada

Howard Cuckle,

MSc, DPhil Professor Faculty of Medicine, Tel Aviv University Tel Aviv, Israel

James E. Davison,

MBChB, PhD Consultant in Paediatric Metabolic Medicine Department of Metabolic Medicine Great Ormond Street Hospital for Children NHS Foundation Trust London, UK

Mina Desai, PhD Associate Professor of Obstetrics & Gynecology David Geffen School of Medicine at UCLA Los Angeles, CA, USA Director of Perinatal Research The Lundquist Institute at Harbor-UCLA Medical Center Torrance, CA, USA

Nadine Girard,

MD, PhD Professor of Neuroradiology Centre de Resonance Magnetique Biologique et Medicale Centre National de la Recherche Scientifique Faculte de Medicine la Timone Université de La Mediterranée Head of Neuroradiology Timone Hospital Aix-Marseille University Marseille, France

Wayne W. Grody, MD, PhD Professor Divisions of Medical Genetics and Molecular Diagnostics Departments of Pathology and Laboratory Medicine, Pediatrics, and Human Genetics University of California Los Angeles School of Medicine UCLA Institute for Society and Genetics Director Molecular Diagnostic Laboratories and Clinical Genomics Center UCLA Medical Center Los Angeles, CA, USA Andrea Gropman,

MD, FAAP, FACMG Professor of Neurology and Pediatrics Chief, Neurogenetics and Neurodevelopmental Disabilities Children’s National Medical Center and the George Washington University School of Medicine Washington DC, USA

Paul J. Hagerman,

Lorraine Dugoff,

MD, PhD Distinguished Professor Department of Biochemistry and Molecular Medicine School of Medicine University of California, Davis Health System Sacramento, CA, USA

Valentine Faure-Bardon

Randi J. Hagerman, MD Distinguished Professor of Pediatrics Endowed Chair in Fragile X Research Medical Director MIND Institute University of California, Davis Health System Sacramento, CA, USA

MD Professor of Obstetrics and Gynecology University of Pennsylvania School of Medicine Chief, Reproductive Genetics Division Department of Obstetrics and Gynecology Hospital of the University of Pennsylvania Philadelphia, PA, USA

Lecturer in Obstetrics and Fetal Medicine Department of Obstetrics and Fetal Medicine Hôpital Necker-Enfants Malades, GH Paris-Centre Université de Paris Paris, France

Steven E. Humphries,

PhD Emeritus Professor of Cardiovascular Genetics Institute of Cardiovascular Science University College London London, UK

Contributors xvii

Marianne H. Hutti, PhD, WHNP-BC, FAANP, FAAN Professor College of Nursing Senior Nurse Scientist University of Kentucky Healthcare – Ambulatory Services University of Kentucky Lexington, KY, USA

Malou A. Lugthart, MD Research Fellow in Fetal Medicine Department of Obstetrics Amsterdam Reproduction and Development Research Institute Amsterdam University Medical Centers University of Amsterdam Amsterdam, The Netherlands

Priya S. Kishnani, MD, FACMGG C.L. and Su Chen Professor of Pediatrics and Professor of Molecular Genetics and Microbiology Duke University School of Medicine Medical Director, YT and Alice Chen Pediatrics Genetics and Genomics Center Division Chief, Medical Genetics Duke Health Durham, NC, USA

Tippi C. MacKenzie,

Bartha Maria Knoppers,

PhD, OC, OQ Canada Research Chair in Law and Medicine Director, Centre of Genomics and Policy Department of Human Genetics Faculty of Medicine McGill University Montreal, Quebec, Canada

Anver Kuliev, MD, PhD Clinical Professor Herbert Wertheim College of Medicine Florida International University Miami, FL Director of Research Reproductive Genetics Innovations Northbrook, IL, USA Marianne Leruez-Ville, MD, PhD Consultant in Medical Virology National Reference Laboratory for Congenital Cytomegalovirus Infections Hôpital Necker-Enfants-Malades, GH Paris Centre Université de Paris Paris, France Brynn Levy,

MSc(Med), PhD, FACMG Professor of Pathology and Cell Biology, Columbia University Medical Center Medical Director, Clinical Cytogenetics Laboratory Co-Director, Laboratory of Personalized Genomic Medicine Vagelos College of Physicians and Surgeons Columbia University Irving Medical Center & the New York Presbyterian Hospital New York, NY, USA

MD Professor of Surgery Co-Director, Center for Maternal-Fetal Precision Medicine University of California, San Francisco San Francisco, CA, USA

Laurence B. McCullough,

PhD Professor Department of Obstetrics and Gynecology Zucker School of Medicine at Hofstra/Northwell Ethics Scholar Department of Obstetrics and Gynecology Lenox Hill Hospital New York, NY, USA

Deborah E. McFadden, MD, FRCPC Clinical Professor Department of Pathology and Laboratory Medicine University of British Columbia Head and Medical Director Department of Pathology and Laboratory Medicine Children’s & Women’s Hospitals of British Columbia Vancouver, BC, Canada Aubrey Milunsky, MD, DSc, FRCP, FACMG, DCH Professor of Obstetrics and Gynecology Tufts University School of Medicine, Boston, MA, USA Formerly Professor of Human Genetics, Pediatrics, Obstetrics and Gynecology, and Pathology Boston University School of Medicine Founder and Co-Director, Center for Human Genetics Cambridge, MA, USA Jeff M. Milunsky,

MD, FACMG Professor of Obstetrics and Gynecology Tufts University School of Medicine Formerly Professor of Genetics and Genomics, and Pediatrics Boston University School of Medicine Co-Director, Center for Human Genetics Director, Clinical Genetics and Molecular Genetics Cambridge, MA, USA

xviii

Contributors

Sara E. Mole, PhD Professor in Molecular Cell Biology UCL Great Ormond Street Institute of Child Health MRC Laboratory of Molecular Cell Biology Genetics and Genomics Medicine Research & Teaching Department Inborn Errors of Metabolism Section University College London London, UK Minh Thu Minh Nguyen, LLM, LLB, BSc Academic Associate Centre of Genomics and Policy McGill University Montreal, Quebec, Canada

Anthony O. Odibo,

MD, MSCE Professor Maternal Fetal Medicine Department of Obstetrics and Gynecology University of South Florida Tampa, FL, USA

John M. Old,

PhD, FRCPath Clinical Scientist Emeritus Reader in Hematology University of Oxford, Oxford, UK

Eva Pajkrt,

MD, PhD Professor of Obstetrics Head of Obstetrics and Fetal Medicine Department of Obstetrics Amsterdam Reproduction and Development Research Institute Amsterdam University Medical Centers University of Amsterdam Amsterdam, The Netherlands

Jennifer M. Puck, MD Professor of Pediatrics Division of Allergy/Immunology and Blood and Marrow Transplantation University of California San Francisco Department of Pediatrics San Francisco, CA, USA Svetlana Rechitsky, PhD Clinical Associate Professor Herbert Wertheim College of Medicine Florida International University Miami, FL, USA President Reproductive Genetic Innovations Northbrook, IL, USA

Wendy P. Robinson, PhD Professor, Department of Medical Genetics University of British Columbia Senior Scientist Child and Family Research Institute Vancouver, BC, Canada David S. Rosenblatt,

MDCM, FCCMG, FRCPSC, FAAP, FACMG Dodd Q. Chu and Family Chair in Medical Genetics Professor Departments of Human Genetics Medicine, Pediatrics, and Biology Faculties of Medicine and Science McGill University Montreal, Quebec, Canada

Michael G. Ross, MD, MPH Distinguished Professor of Obstetrics and Gynecology and Public Health Geffen School of Medicine at University of California Los Angeles Fielding School of Public Health at UCLA Los Angeles, CA, USA Co-Director Institute for Women’ and Children’s Health The Lundquist Institute at Harbor-UCLA Medical Center Torrance, CA, USA Marisa E. Schwab, MD Surgery Resident The Center for Maternal-Fetal Precision Medicine and Department of Surgery University of California, San Francisco San Francisco, CA, USA Alireza A. Shamshirsaz Associate Professor of Surgery, Obstetrics and Gynecology Departments of Obstetrics and Gynecology and Surgery Baylor College of Medicine Chief, Division of Fetal Therapy and Surgery Co-Chief, Maternal Fetal Surgery Section Texas Children’s Fetal Center Texas Children’s Hospital Houston, TX, USA

Lee P. Shulman,

MD, FACMG, FACOG The Anna Ross Lapham Professor in Obstetrics and Gynecology Feinberg School of Medicine of Northwestern University Medical Director, Insight Medical Genetics Medical Director, Reproductive Genetic Innovations Chicago, IL, USA

Contributors xix

Andrea Superti-Furga,

MD

Professor of Pediatrics University of Lausanne Head, Division of Genetic Medicine Lausanne University Hospital Lausanne, Switzerland

Jan Traeger-Synodinos,

DPhil (Oxon), ErCLG Professor of Genetics Director of the Laboratory of Medical Genetics National and Kapodistrian University of Athens, Greece

Sheila Unger,

MD, FRCPC Chief MER1 Division of Genetic Medicine Lausanne University Hospital Lausanne, Switzerland

Ignatia B. Van den Veyver,

MD Professor of Obstetrics and Gynecology and Molecular and Human Genetics Baylor College of Medicine; Director of Clinical Prenatal Genetics Texas Children’s Hospital Houston, TX, USA

Daniel L. Van Dyke,

PhD Professor of Laboratory Medicine and Pathology Mayo Medical School and Mayo Clinic Cytogenetics Laboratory Rochester, MN, USA

Liesbeth van Leeuwen,

MD, PhD Associate Professor Department of Obstetrics Amsterdam Reproduction and Development Research Institute Amsterdam University Medical Centers University of Amsterdam Amsterdam, The Netherlands

Yves G. Ville,

MD Professor of Obstetrics and Fetal Medicine Head of Department of Obstetrics and Fetal Medicine Hôpital Necker-Enfants Malades, GH Paris-Centre Université de Paris Paris, France

Gerard Vockley,

MD, PhD Professor of Pediatrics and Human Genetics Cleveland Family Endowed Professor of Pediatric Research Department of Pediatrics University of Pittsburgh School of Medicine and Graduate School of Public Health Chief, Division of Medical Genetics Director, Center for Rare Disease Therapy Children’s Hospital of Pittsburgh Pittsburgh, PA, USA

Ronald J.A. Wanders University of Amsterdam, Academic Medical Center Departments of Clinical Chemistry and Pediatrics Emma Children’s Hospital Laboratory of Genetic Metabolic Diseases Amsterdam, The Netherlands

Hans R. Waterham,

PhD Professor, Functional Genetics of Metabolic Diseases Departments of Laboratory Medicine and Pediatrics University of Amsterdam Academic Medical Center Laboratory for Genetic Metabolic Diseases Amsterdam, The Netherlands

David Watkins,

PhD Research Associate Department of Human Genetics McGill University Scientist Division of Medical Genetics, Department of Specialized Medicine McGill University Health Centre Montreal, Quebec, Canada

Bryan G. Winchester,

MA, PhD Emeritus Professor of Biochemistry ULC Great Ormond Street Institute of Child Health University College London London, UK

Aline Wolter, MD Justus-Liebig-University, Giessen Department of Obstetrics and Gynecology Division of Prenatal Medicine and Fetal Therapy University Hospital Giessen, Germany Ellen Wright Clayton,

MD, JD Craig-Weaver Professor of Pediatrics Professor of Law Center for Biomedical Ethics and Society Vanderbilt University Nashville, TN, USA

1

Genetic Counseling: Preconception, Prenatal, and Perinatal Aubrey Milunsky 1,2 and Jeff M. Milunsky 1,2 1 2

Center for Human Genetics, Cambridge, MA, USA Tufts University School of Medicine, Boston, MA, USA

The time is fast approaching when virtually all the culprit genes and their mutations for >7,000 rare monogenic disorders1 will be known. Thus far, causal single genes and their mutations have been determined for 5,673 genetic disorders,2 enabling preimplantation genetic testing or prenatal genetic diagnosis. These advances using chromosomal microarrays, whole-exome sequencing and even whole-genome sequencing together with fetal imaging and noninvasive prenatal testing, expand the era in which all couples have the option of avoiding or preventing having children with irreversible, irremediable, crippling, or lethal monogenic disorders. Primary care physicians, and those in all medical specialties, will need to inform their patients of this key option. This imperative is already partly in current practice. Missing is the requirement of physicians to request and obtain the precise name of the genetic disorder in question or an existing DNA report on a family member, for prospective parents to benefit from available options. Increasingly, couples are seeking prenatal diagnosis for adult-onset genetic disorders in which mutations have been determined. Huntington disease prenatal diagnosis has been in the vanguard for many years, but now there are requests for adult-onset dominantly transmissible disorders including breast and other malignancies,

frontotemporal dementia, neurodegenerative disorders, and cardiomyopathies. The remarkable advances in genetics provide a cogent need to confer and refer. Physicians should not invite legal purview for a failure to inform, offer, refer, or provide genetic testing. In context, couples at risk for having progeny with abnormalities expect to be informed about their risks and options, optimally during preconception counseling. Their concerns are serious, given the significant contribution of genetic disorders to morbidity and mortality in children and adults.

The burden of genetic disorders and congenital malformations A conservative estimate for the world population prevalence of rare diseases (71.9–80 percent considered as genetic) is 3.5–5.9 percent, equating to 263–446 million individuals affected at any point in time.3, 4 In India, which has a quarter of the world’s neonatal deaths (an estimated 753,000 in 2013), about 9 percent were due to congenital anomalies.5 An estimated 7.9 million infants worldwide are born each year with a major congenital malformation according to a report in 2013.6 The likelihood of having a child with a congenital malformation varies from 2 to 10 percent7 due to multiple factors

Genetic Disorders and the Fetus: Diagnosis, Prevention, and Treatment, Eighth Edition. Edited by Aubrey Milunsky and Jeff M. Milunsky. © 2021 Aubrey Milunsky and Jeff M. Milunsky. Published 2021 by John Wiley & Sons Ltd.

1

2

Genetic Disorders and the Fetus

that complicate efforts to accurately diagnose and determine the incidence or prevalence of congenital anomalies or genetic disorders. Box 1.1 lists the majority of known etiologic categories, discussed below, which help explain sometimes striking differences among major studies. It is almost impossible to account for all these potentially confounding factors in a study, and rarely has any one study come close. Of the >7,000 rare genetic disorders, about 1 in 12 to 1 in 16 individuals are affected,1 aware or unaware. Given a world population of 7.6 billion, an estimated 473 million are likely to have a rare disease.1

More than 4,331 genes with phenotype-causing mutations have been identified, including 6,739 phenotypes with a known molecular basis.2 Severe intellectual disability is considered to be largely genetic in origin8, 9 with a global prevalence between 0.5 and 1.0 percent.10 Despite continuing progress in the discovery of genes causally related to neurodevelopmental delay,11 in less than 40 percent of cases is there a definitive recognition of cause. The European Organization for Rare Diseases maintained that about 30 percent of all patients with a rare disease died before the age of 5 years.12 In the United States in 2013–2014,

Box 1.1 Factors that influence estimates of the incidence or prevalence in the newborn of a congenital malformation (CM) or genetic disorder Availability and use of expertise in prenatal diagnostic ultrasound and MRI • Accuracy of diagnosis • Age at diagnosis • Case selection, bias, and ascertainment • Congenital hypothyroidism • Consanguinity • Definitions of major and minor congenital anomalies • Diagnostic DNA analysis • Duration of follow-up • Economic level in developed or developing world • Environmental toxins • Family history • Frequency, inclusion, and exclusion of stillbirths, fetal deaths, and elective pregnancy termination • Frequency of certain infectious diseases • Frequency of de novo gene mutations • History of recurrent spontaneous abortion • In vitro fertilization • Incidence and severity of prematurity • Infertility • Intracytoplasmic sperm injection • Later manifestation or onset of disorder • Maternal age • Maternal alcohol abuse • Maternal diabetes and gestational diabetes • Maternal diet • Maternal epilepsy, lupus erythematosus and other illnesses •

Maternal fever or use of hot tub in the first 6 weeks of pregnancy • Maternal folic acid supplementation • Maternal grandmother’s age • Maternal obesity • Maternal serum screening for chromosome abnormalities • Maternal smoking • Maternal-specific susceptibility genes • Maternal use of medication • Mortality rates decreasing • Multiple pregnancy rate • Necropsy • Noninvasive prenatal testing using cell-free fetal DNA for chromosomal abnormalities and monogenic disorders • Parent with a congenital abnormality or genetic disorder • Paternal age • Previous affected child • Previous maternal immunization/vaccination • Season of the year • Training and expertise in examination of newborns • Use of chromosomal analysis • Use of chromosomal microarray • Use of whole-exome sequencing • Use of whole-genome sequencing • Use of death certificates • Use of registry data •

CHAPTER 1

Genetic Counseling: Preconception, Prenatal, and Perinatal 3

congenital malformations, deformations, and chromosomal abnormalities accounted for the most infant deaths – 4,746 (20.4 percent) out of 23,215 – in any category of causation.13

Incidence and prevalence of genetic disorders and congenital malformations Estimates of aneuploidy in oocytes and sperm reach 25 percent and 3–4 percent, respectively.14, 15 Estimates, especially for oocytes, vary widely (see Chapter 2). The effect of maternal age, among other factors, is important. At 25 years, early thirties, and >40 years of age, the rate of aneuploidy approximates 5 percent, 10–25 percent, and 50 percent, respectively.15–19 Estimates of aneuploidy and structural chromosomal abnormalities in sperm vary from 7 to 14 percent.20 Not surprisingly, then, about one in 13 conceptions results in a chromosomally abnormal conceptus,21 while about 50 percent of first-trimester spontaneous abortions are associated with chromosomal anomalies.22 One study of blastocysts revealed that 56.6 percent were aneuploid. Moreover, these blastocysts produced in vitro from women of advanced maternal age also revealed mosaicism in 69.2 percent.23 Similar results have been reported by others.24 Clinically significant chromosomal defects occur in 0.65 percent of all births; an additional 0.2 percent of babies are born with balanced structural chromosome rearrangements that have implications for reproduction later in life (see Chapters 11 and 13). Between 5.6 and 11.5 percent of stillbirths and neonatal deaths have chromosomal defects.25 Congenital malformations with obvious structural defects are found in about 2 percent of all births.26 This was the figure in Spain among 710,815 livebirths,27 with 2.25 percent in Liberia,28 2.03 percent in India,29 and 2.53 percent among newborn males in Norway.30 The Mainz Birth Defects Registry in Germany in the 1990–1998 period reported a 6.9 percent frequency of major malformations among 30,940 livebirths, stillbirths, and abortions.31 Pooled data from 12 US population-based birth defects surveillance systems, which included 13.5 million livebirths (1999–2007), revealed that American Indians/Alaska natives had a ≥50 percent greater prevalence for seven congenital malformations (including anotia or microtia, cleft lip, trisomy 18, encephalocele, limb-reduction

defect).32 Factors that had an impact on the incidence/prevalence of congenital malformations are discussed later. Over 25,500 entries for genetic disorders and traits have been catalogued.2 Estimates based on 1 million consecutive livebirths in Canada suggested a monogenic disease in 3.6 in 1,000, consisting of autosomal dominant (1.4 in 1,000), autosomal recessive (1.7 in 1,000), and X-linked recessive disorders (0.5 in 1,000).33 Baseline birth prevalence of rare single-gene disorders for multiple countries are shown in Figure 1.1, which highlights the contribution of consanguinity-associated disorders.34 Polygenic disorders occurred at a rate of 46.4 in 1,000 (Table 1.1). A key study of homozygosity in consanguineous patients with an autosomal recessive disease showed that, on average, 11 percent of their genomes were homozygous.35 Each affected individual had 20 homozygous segments exceeding 3 cM. At least 3–4 percent of all births are associated with a major congenital defect, intellectual disability, or a genetic disorder, a rate that doubles by 7–8 years of age, given later-appearing and/or later-diagnosed genetic disorders.36, 37 If all congenital defects are considered, Baird et al.33 estimated that 7.9 percent of liveborn individuals have some type of genetic disorder by about 25 years of age. These estimates are likely to be very low given, for example, the frequency of undetected defects such as bicuspid aortic valves that occur in 1–2 percent of the population.38 The bicuspid aortic valve is the most common congenital cardiac malformation and in the final analysis may cause higher mortality and morbidity rates than all other congenital cardiac defects.39 About 27 percent suffer cardiovascular complications requiring surgery.40, 41 Mitral valve prolapse affects 2–3 percent of the general population, involving more than 176 million people worldwide.42 A Canadian study of 107,559 patients with congenital heart disease reported a prevalence of 8.21 per 1,000 livebirths, rising to an overall prevalence of 13.11 per 1,000 in adults.43 The authors concluded that adults now account for some two-thirds of the prevalence of congenital heart disease. Categorical examples of factors associated with an increased risk of congenital heart disease or malformations in the fetus are shown in Box 1.1. A metropolitan Atlanta study (1998–2005) showed an overall

4

Genetic Disorders and the Fetus

22 20 Consang-associated

Rate/1,000 births

18 16

Genetic type unknown

14

Recessive

12 10

X-linked

8 Early onset dominant

6 4 2 0

EMR

AFR

SEAR

EUR

WPR

AMR

World W Europe

Figure 1.1 Total baseline birth prevalence of rare single-gene disorders by World Health Organization (WHO) region, highlighting the important contribution of consanguinity to monogenic disorders. Source: Blencowe et al. 2018.34 Reproduced with permission from Springer.

Table 1.1 The frequencies of genetic disorders in 1,169,873 births, 1952–198334 . Rate per million

Total births

livebirths

(percent)

Dominant

1,395.4

0.14

Recessive

1,665.3

0.17

X-linked

532.4

0.05

Chromosomal

1,845.4

0.18

Multifactorial

46,582.6

4.64

Category A

Genetic unknown

1,164.2

0.12

Total

53,175.3

5.32 a

B All congenital anomalies 740–759 b

52,808.2

5.28

Congenital anomalies with genetic etiology

26,584.2

2.66

79,399.3

7.94

(included in section A) C Disorders in section A plus those congenital anomalies not already included a Sum

is not exact owing to rounding.

b International

Classification of Disease numbers.

Source: Blencowe et al. 2018.34 With permission from Elsevier.

prevalence of 81.4 per 10,000 for congenital heart disease among 398,140 livebirths,44 similar to a Belgian study of 111,225 live and stillborn infants ≥26 weeks of gestation with an incidence of 0.83 percent, chromosome abnormalities excluded.45 A EUROCAT registry study found an increasing prevalence of severe congenital heart defects (single ventricle, atrioventricular septal defects, and tetralogy of Fallot) possibly due to increasing

obesity and diabetes.46 In a study of 8,760 patients with autism spectrum disorders and 26,280 controls, a statistically significant increase in the odds of concurrent congenital heart disease (odds ratio [OR] 1.32) was noted.47 Atrial septal defects and ventricular septal defects were most common. Incidence/prevalence rates of congenital defects are directly influenced by when and how diagnoses are made. Highlighting the importance of how

CHAPTER 1

Genetic Counseling: Preconception, Prenatal, and Perinatal 5

early a diagnosis is made after birth, the use of echocardiography, and the stratification of severity of congenital heart defects, Hoffman and Kaplan48 clarified how different studies reported the incidence of congenital heart defects, varying from 4 in 1,000 to 50 in 1,000 livebirths. They reported an incidence of moderate and severe forms of congenital heart disease in about 6 in 1,000 livebirths, a figure that would rise to at least 19 in 1,000 livebirths if the potentially serious bicuspid aortic valve is included. They noted that if all forms of congenital heart disease (including tiny muscular ventricular septal defects) are considered, the incidence increases to 75 in 1,000 livebirths. The newer genetic technologies, including chromosomal microarray, whole-exome sequencing, next-generation sequencing, and whole-genome sequencing, have helped unravel the causes of an increasing number of isolated or syndromic congenital heart defects.49, 50 Identified genetic causes include monogenic disorders in 3–5 percent of cases, chromosomal abnormalities in 8–10 percent, and copy number variants in 3–25 percent of syndromic and 3–10 percent of isolated congenital heart defects.49, 51 A next-generation sequencing study indicated that 8 percent and 2 percent of cases were due to de novo autosomal dominant and autosomal recessive pathogenic variants, respectively.52 Pregestational diabetes in 775 of 31,007 women was statistically significantly associated with sacral agenesis (OR 80.2), holoprosencephaly (OR 13.1), limb reduction defects (OR 10.1), heterotaxy (12.3), and severe congenital heart defects (OR 10.5–14.9).53 Maternal obesity is associated with an increased risk of congenital malformations.54–65 The greater the maternal body mass index (BMI), the higher the risk, especially for congenital heart defects,59, 60, 62, 65 with significant odds ratios between 2.06 and 3.5. In a population-based case–control study, excluding women with preexisting diabetes, Block et al.66 compared the risks of selected congenital defects among obese women with those of average-weight women. They noted significant odds ratios for spina bifida (3.5), omphalocele (3.3), heart defects (2.0), and multiple anomalies (2.0). A Swedish study focused on 1,243,957 liveborn singletons and noted 3.5 percent with at least one major congenital abnormality.64

These authors used maternal BMI to estimate risks by weight. The risk of having a child with a congenital malformation rose steadily with increasing BMI from 3.5 percent (overweight) to 4.7 percent (BMI ≥40). Our own67, 68 and other studies69 have implicated the prediabetic state or gestational diabetes as contributing to or causing the congenital anomalies in the offspring of obese women. In this context, preconception bariatric surgery seems not to reduce the risks of congenital anomalies.61, 70–72 It appears that folic acid supplementation attenuates but does not eliminate the risk of spina bifida when associated with diabetes mellitus73 or obesity74 (see Chapter 10). In contrast, markedly underweight women reportedly have a 3.2-fold increased risk of having offspring with gastroschisis,74 in all likelihood due to smoking and other acquired exposures.75, 76 Indeed, a study of 173,687 malformed infants and 11.7 million unaffected controls, when focused on maternal smoking, yielded significant odds ratios up to 1.5 for a wide range of major congenital malformations in the offspring of smoking mothers.77 Young nulliparous women have an increased risk of bearing a child with gastroschisis, those between 12 and 15 years of age having a more than fourfold increased risk.78 A Californian population-based study (1995–2012) recorded a prevalence for gastroschisis of 2.7 cases per 10,000 livebirths.75 The surveillance system of the National Network of Congenital Anomalies of Argentina reported a 2009–2016 study of 1,663,610 births with 702 born with limb reduction defects.79 The prevalence was 4.22/10,000 births. In 15,094 stillbirths, the prevalence rose to 30.80/10,000. A Chinese study of 223 newborn deaths in a neonatal intensive care unit noted that 44 (19.7 percent) had a confirmed genetic disorder.80 The National Perinatal Epidemiology Centre in Ireland in a study of fatal fetal anomalies recorded 2,638 perinatal deaths, 939 (36 percent) having a congenital anomaly, 43 percent of which were chromosomal.81 More than a single anomaly was noted in 36 percent (333 of 938) of their cases. These numbers led to a significant genetic disease burden and have accounted for 28–40 percent of hospital admissions in North America, Canada, and England.82–84 Notwithstanding their frequency, the causes of about 60 percent of congenital malformations remain obscure.85, 86

6

Genetic Disorders and the Fetus

The effect of folic acid supplementation, via tablet or food fortification, on the prevalence of neural tube defects (NTDs), is now well known to reduce the frequency of NTDs by up to 70 percent87, 88 (see Chapter 10). A Canadian study focused on the effect of supplementation on the prevalence of open NTDs among 336,963 women. The authors reported that the prevalence of open NTDs declined from 1.13 in 1,000 pregnancies before fortification to 0.58 in 1,000 pregnancies thereafter.89 In a population-based cohort study by the Metropolitan Atlanta Congenital Defects Program, the risk of congenital malformations was assessed among 264,392 infants with known gestational ages, born between 1989 and 1995. Premature infants (16

Immune system Susceptibility to infection

100

Juvenile rheumatoid-like arthritis

1.2

Gastrointestinal Congenital defects of the gastrointestinal tract

6

Celiac disease

5.4

Dysphagia

55

Endocrine/metabolic Overweight/obesity

23–70

Hypothyroidism

50

Diabetes mellitus

1.4–10.6

Hyperthyroidism

1–3

Ophthalmologic Eye disorders a

80

Cataract

17–29

Keratoconus

8–10

Hematologic/oncologic Leukemia

2–3 (>20-fold excess)

Testicular cancer

Standardized incidence ratio of 2.9

Transient myeloproliferative disorder

50 years of age had associated risks of fetal death almost twice that of younger fathers.426 A Swiss population study found that the proportion of younger fathers was uniformly different between those with and without Down syndrome offspring. Young fathers had an almost twofold increased odds for siring a child with trisomy 21.427 The authors stated the need for confirmation of their findings. Paternal age should garner more attention during genetic counseling,428 especially with the availability of molecular analysis of multiple genes susceptible to de novo mutations in both noninvasive prenatal testing (see Chapter 8) and prenatal diagnosis (see Chapter 14).

A previous fetus or child with a genetic disorder A genetic evaluation and counseling are usually indicated when a previous fetus or child has or had a genetic disorder, unless the matter is straightforward (e.g. previous trisomy 21) and the obstetrician is well informed. Careful inquiry should be made about the health status of a previous child. Failure or delay in the diagnosis of a monogenic disorder leaves the parents without the option of prenatal diagnosis in a subsequent pregnancy. In addition, it deprives them of the option of preimplantation genetic testing for those disorders with known mutations. Failure to make an early diagnosis of a genetic disorder during the first 5 years of life is common. For example, the Rotterdam Clinical Genetics Group reported that 50 percent of children affected by neurofibromatosis had been treated for related symptoms before a specific diagnosis had been made.429 Such delay has become

problematic given that the NF1 gene and genes for many other monogenic disorders are routinely sequenced for a precise diagnosis. Frequently, distressed parents will select a different physician for a subsequent pregnancy and a new or more recent insight may shed light on the cause of the previous disorder. For example, confined placental mosaicism (see Chapter 4) may now serve to explain the discrepancy between reported chromosomal findings at the time of CVS and fetal tissues obtained at elective abortion. Confined placental mosaicism may also be associated with intrauterine growth restriction (see Chapter 4), requiring serial ultrasounds during the pregnancy. Given the heterogeneous nature of genetic disease, being alert to alternative mechanisms of causation will on occasion be rewarding. For example, during a consultation with a patient who had previously delivered a child with the autosomal recessive Meckel–Gruber syndrome, preparatory discussions about establishing the specific mutation from each parent could reveal that the father is not a carrier of a mutation in the culprit gene. Although nonpaternity is more likely, a judicious approach would also include consideration of uniparental disomy.430, 431 This mode of inheritance, in which an offspring can inherit two copies – part or all of a chromosome from one parent and no copy from the other parent – has been seen in a number of disorders, including Prader–Willi syndrome and Angelman syndrome (see discussion later and Chapter 14). About 25 percent of cases of Prader–Willi syndrome are caused by maternal uniparental disomy.432 Involvement of chromosomes 7, 11, 14, and 15 have been notable. Uniparental disomy is caused primarily by meiotic nondisjunction events and followed by trisomy or monosomy “rescue.” Most cases described have been associated with advanced maternal age and have been detected primarily in the process of prenatal genetic studies.433, 434 Recognition of the molecular basis of a disorder from which a previous child died may provide a couple with an opportunity for prenatal diagnosis in a subsequent planned pregnancy. A caveat would be the availability of analyzable tissue from the deceased child. In the recent past this was mostly not done, but with the escalation of new discoveries in genetics, tissues should now be frozen for potential future DNA analysis. The establishment

CHAPTER 1

Genetic Counseling: Preconception, Prenatal, and Perinatal 29

of the molecular basis of recognized syndromes, previously undetectable prenatally, now provides new opportunities for couples seeking prenatal diagnosis. Examples abound and include some of the craniosynostosis syndromes, certain skeletal dysplasias, and many other disorders. In one of our cases, a father with metaphyseal dysplasia of Schmid, troubled by the indignities and hurts of growing up with severe short stature, elected prenatal diagnosis at a preconception visit. Subsequent mutation analysis of conceived twins yielded a normal prenatal diagnosis result confirmed postnatally.435 Heterogeneity and pleiotropism also require consideration in the context of a previous child’s disorder and anticipation of future prenatal diagnosis. For example, a previous child with tuberous sclerosis or a fetus with a cardiac rhabdomyoma would prompt molecular analysis of the TSC1 and TSC2 genes for more precise future prenatal diagnosis.436

A parent with a genetic disorder Physicians are now advised to determine whether a culprit gene has been found for a specific genetic disorder under discussion, since prenatal diagnosis would then be available for that couple or their children. Adult-onset genetic disorders (breast/ovarian cancer, colon cancer, hypertrophic cardiomyopathy, long QT syndrome) serve as examples where prenatal diagnosis is an option. The long-established prenatal diagnoses for both presymptomatic and symptomatic neurodegenerative disorders437 continue to be expanded to include disorders such as amyotrophic lateral sclerosis and frontotemporal dementia by analysis of the C9orf72 gene.438 In prenatal diagnosis discussions for all adult-onset disorders, there is a natural focus on the tortured questions of personal existence and self-extinction. One example is that of a young father with CADASIL (cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy) who, faced with our prenatal diagnosis of this disorder, by mutation analysis of the Notch3 gene, with his wife, elected termination.439 Mutation analysis in a subsequent pregnancy assured an unaffected fetus.440 These consultations may invoke deep personal emotional conflict, especially when pleiomorphic genes are concerned. For example, a parent with tuberous sclerosis and normal intelligence could

not be certain that an affected child would not have intellectual disability. This was especially evident in our series of 50 couples having prenatal diagnosis for tuberous sclerosis.436 Discovery of fetal cardiac rhabdomyoma led to sequencing of both the TSC1 and TSC2 genes in the fetus and diagnosis in one of the asymptomatic parents. Parental decisions are neither simple nor predictable. In a UK study441 of 644 deaf individuals and 143 with hearing impairment, 2 percent opined that they would prefer to have deaf children and would consider an elective abortion if the fetus was found to be hearing! Prospective mothers with insulin-dependent diabetes mellitus (IDDM) could find their disorder harder to control during pregnancy. Diabetes should be well controlled before pregnancy. The better the control, the lower the risk of having a child with congenital defects.442, 443 An Australian study noted that with good preconception care of type 1 IDDM, the major congenital malformation rate decreased from a high of 14 percent to 2.2 percent.444 Notwithstanding extant knowledge about IDDM and pregnancy, a report of 273 women noted rates of stillbirth (1.85 percent), perinatal mortality (2.78 percent), and congenital anomalies (6 percent).445 An important Stockholm study of 1,089 stillbirths usefully separated causes in preterm and term/post-term births.446 Infection and intrauterine growth restriction/placental insufficiency accounted for over 44 percent of cases in about equal proportion. The genetics of diabetes is complex with multiple types, both polygenic, multifactorial, syndromic, and monogenic in origin. The polygenic type 1 diabetes (T1DM) and type 2 diabetes (T2DM) have over 40 and 90 genes implicated, respectively. Between 1 and 5 percent of diabetes is monogenic and symptoms overlap with T1DM and T2DM diabetes.447, 448 Affected monogenic type patients mostly do not have islet autoantibodies, often have endogenous insulin production, and are frequently misdiagnosed.449, 450 Both T2DM and monogenic diabetes are often not insulin-dependent, have a family history of diabetes, and can occur in the young. Usually, insulin resistance does not occur, nor does acanthosis nigricans in monogenic diabetics, who are mostly not obese.449 Diabetes diagnosed in the first year of life is monogenic and due to KATP channel mutations.451 There are multiple types of monogenic autosomal

30 Genetic Disorders and the Fetus

dominant maturity-onset diabetes of the young (MODY), four subtypes predominating with mutations in HNFIA (52 percent), GCK (32 percent), HNF4A (10 percent), and HNF1B (6 percent).452 A precise preconception molecular diagnosis is important so as to direct appropriate treatment. No pharmacologic treatment is indicated for the GCK-MODY type, low dose sulfonylureas are prescribed for HNF1A-MODY and HNF4A-MODY, with high-dose sulfonylureas for KATP channel-related diabetes.451 Pregestational T1DM and T2DM are associated with poorer pregnancy outcomes, including up to a fourfold higher rate of perinatal mortality.453 The poorer glycemic control at the time of conception and the first trimester, the higher the frequency of stillbirths, congenital abnormalities, perinatal morbidity and mortality, macrosomia, dystocia in labor, and maternal mortality.454–458 Obesity, with its burden of obstetric complications and congenital anomalies457 (as discussed earlier), compounds all the problems in the diabetic mother. Pregnant women with the chronic multifactorial autoimmune disease systemic lupus erythematosus (SLE) face a host of complications. This disorder, with its predilection for women of childbearing age, is more prevalent in non-white populations and is characterized by involvement that includes renal, cardiovascular, musculoskeletal, neurological, rheumatological, and cutaneous systems.459 Adverse pregnancy outcomes include fetal death, preterm births, intrauterine growth restriction, and neonatal lupus.460 Women with anti-Ro/anti-La antibodies, the latter being specific for the diagnosis of SLE and Sjögren syndrome,461 can be asymptomatic. Anti-Ro antibodies may precede the clinical manifestations of SLE by an average of 3.6 years.462 Note, however, these antibodies are found in up to 3 percent of the general population.463 The prime consequences of having anti-Ro antibodies is the risk of fetal/neonatal heart block and neonatal lupus. In a study of 325 children with second- or third-degree heart block, the overall mortality rate was 17.5 percent. Death in utero occurred in 6 percent.464 The risk of offspring being born with congenital heart block to a mother with anti-Ro antibodies is between 0.2 and 2.0 percent, but 15–20 percent if there has been a previously affected fetus or neonate.464, 465 After

two affected pregnancies, the subsequent pregnancy risk is 50 percent.466 Complex therapeutic considerations include fluorinated glucocorticoids (dexamethasome and betamethasome) and maternal fetal echocardiography monitoring.467, 468 Neonatal lupus with congenital heart block will usually require pacemaker implantation.469, 470 For mothers with a previous affected pregnancy, hydroxychloroquine has been recommended as a pre-emptive treatment.471, 472 Fortunately, only a third of mothers carrying fetuses with complete heart block have an identified autoimmune disorder such as lupus or Sjögren disease.473 Certain genetic disorders may threaten maternal and fetal health in pregnancy and are discussed in detail in Chapter 31.

A history of infertility Beyond the issues of paternal age discussed earlier, there is the evidence that structural chromosomal abnormalities, which occur in 0.25 percent of births, more frequently have their origin in paternal chromosomes. In a 2006 report, 72 percent of de novo unbalanced chromosomal rearrangements were of paternal origin.474 The likelihood of having a translocation doubled every 10 years after the age of 25.475 An American Cancer Society Study of 2,532 cases of hematological cancers noted that men over 35 had a 63 percent higher risk of having affected offspring when compared with those under 25.476 A small, but statistically significant increased risk of nonchromosomal congenital malformations associated with advanced paternal age was reported by the National Birth Defects Prevention Study.477 Malformations included were cleft lip, diaphragmatic hernia, right ventricular outflow tract obstruction, and pulmonary stenosis. About 10 percent of couples have infertility. A World Health Organization multicenter study concluded that the problem appeared predominantly in males in 20 percent of cases, predominantly in females in 38 percent, and in both partners in 27 percent. In the remaining 15 percent of cases, no definitive cause for the infertility was identified.478 Care should be exercised in the preconception counseling of a couple with a history of infertility. In the absence of a recognizable cause, karyotyping of both is recommended. Unrecognized spontaneous abortions may have occurred without the patient’s awareness, caused by overt structural

CHAPTER 1

Genetic Counseling: Preconception, Prenatal, and Perinatal 31

chromosome rearrangements or microdeletions or duplications (see Chapters 11 and 13). Microarrays performed after routine cytogenetics on products of conception in 2,389 cases revealed significant copy number changes or whole-genome uniparental disomy in 1.6 percent and 0.4 percent of cases, respectively.479 A study of 1,300 infertile men revealed chromosomal abnormalities in 10.6 percent and Y-microdeletions in 4.0 percent.480 Recognized habitual abortion due to the same causes would also require cytogenetic analysis. Such studies may reveal a parent (rarely both) with a chromosomal rearrangement with significant risks for bearing a child with intellectual disability and/or malformations, who could benefit from prenatal or preimplantation diagnosis. Other examples of disorders characteristically associated with recurrent pregnancy loss or infertility include premature ovarian failure in fragile X syndrome carriers (see Chapter 16), and the X-linked disorders steroid sulfatase deficiency481 and incontinentia pigmenti.482 Thrombophilia as a significant cause remains uncertain.483, 484 In about 8 percent of women experiencing recurrent abortion a mutation in the SYCP3 gene (which encodes an essential component of the synaptonemal complex, key to the interaction between homologous chromosomes) was noted.485 An extensive list of genes related to premature ovarian failure have been recognized,486 especially noteworthy in a highly consanguineous population.487 Consequently, next-generation sequencing488 or whole-exome sequencing,489–492 cost issues aside, would be indicated. Although the investigation to determine the cause of male or female infertility can be extensive, several observations are pertinent here. We recognized that congenital bilateral absence of the vas deferens (CBAVD),493 which occurs in 1–2 percent of infertile males, is primarily a genital form of CF (see Chapter 15). Men with CBAVD494 should have CF gene analysis (sequencing, poly T variant analysis, deletion analysis). A meta-analysis concluded that among CBAVD patients, 78 percent had one recognizable CFTR gene mutation whereas 46 percent were noted to have two mutations.495 The mutation detection rate is likely to exceed 92 percent including large gene rearrangements.496 Of interest is the observation of Traystman et al.497 that CF carriers may be at higher risk for infertility

than the population at large. Men who test negative for a CFTR mutation should have the ADGRG2 gene on the X chromosome sequenced.498, 499 Some patients with CBAVD (21 percent in one study500 ) also have renal malformations. These patients may have a normal sweat test and thus far no recognizable mutations in the CF gene.500, 501 Renal ultrasound studies are recommended in all patients with CBAVD who have normal CFTR analyses. The partner of a male with CBAVD and a recognized mutation(s), after gene analysis, should routinely be offered sequencing and deletion analysis of the CFTR gene. Such couples frequently consider epididymal sperm aspiration,502, 503 with pregnancy induced by IVF. Precise prenatal and/or preimplantation genetic testing can be achieved only if specific mutations have been recognized. Significant male infertility is mainly associated with XXY males (see Chapter 12), autosomal translocations, Kallman syndrome, Ymicrodeletions, autosomal inversions, CBAVD, mixed gonadal dysgenesis, and X-linked and autosomal gene mutations.504 We reported a 28-year-old with azoospermia and bilateral congenital cataracts associated with a contiguous deletion including the Nance–Horan gene at Xp23.13 and implicating the SCML1 gene.505 The global prevalence of Yq microdeletions approximates 7.5 percent in infertile males.506 Genes including DAZ (“deleted in azoospermia”), YRRM (Y chromosome RNA recognition motif),507, 508 and others may be deleted singly or together in the region of Yq11.23.509 Couples must be informed that male offspring of men with these interstitial deletions in the Y chromosome will have the same structural chromosome defect. The female partner of the male undergoing intracytoplasmic sperm injection (ICSI) needs explanations about procedures and medications for her that are not risk free. Patients should realize that ICSI followed by IVF is likely to achieve pregnancy rates between 20 and 24 percent,510 a success rate not very different from the approximately 30 percent rate in a single cycle after natural intercourse at the time of ovulation.510 Pregnancy follow-up data from cases culled from 35 different programs reported in a European survey511 and a major American study of 578 newborns showed no increased occurrence of congenital malformations.214 However, a statistically significant increase in sex chromosome

32 Genetic Disorders and the Fetus

defects has been observed.512 Prenatal diagnosis is recommended in all pregnancies following ICSI. Even “balanced” reciprocal translocations in males may be associated with the arrest of spermatogenesis and resultant azoospermia.513 In one series of 150 infertile men with oligospermia or azoospermia, an abnormal karyotype was found in 10.6 percent (16/180), 5.3 percent (8/150) had an AZF-c deletion, and 9.3 percent (14/150) had at least a single CF gene mutation.514 This study revealed a genetic abnormality in 36/150 (24 percent) of men with oligospermia or azoospermia. A Turkish study of 1,696 males with primary infertility showed 8.4 percent with a chromosomal abnormality and 2.7 percent with a Y-chromosome microdeletion.515 Rarer disorders may need to be considered in the quest to determine the cause of infertility including, for example, the blepharophimosis, ptosis, epicanthus inversus syndrome, which may respond to treatment.516 In a study of 75,784 women to determine all-cause and cause-specific mortality, those with infertility had a 10 percent increased risk of death from any cause.517 Death from breast cancer was more than doubled. In a major prospective Danish study, 3,356 women who had children born after frozen embryo transfer were compared with 910,291 fertile women. The incidence rate of childhood cancer was 17.5 per 100,000 for children born to fertile women, and 44.4 per 100,000 in children born after the use of frozen embryos.518 The statistically significant increased risk was primarily leukemia and sympathetic nervous system tumors. The cause(s) remain unknown. A US study did not find a significant association, but had a shorter follow-up period (30 percent complained of anxiety, depression, and headaches.533 Between 20 and 30 percent of carriers experience irregular or absent menses due to primary ovarian insufficiency.534 This latter recognition during routine obstetric care often serves as an alert to check fragile X syndrome carrier status. We have also seen instances where recognition of carrier status has led to reversal of a putative diagnosis of parkinsonism or early dementia, instead of an actual diagnosis of the fragile X tremor ataxia syndrome manifesting in a grandfather over 60 years of age (see Chapter 16). Carrier status for women with a family history of hemophilia A or B cannot be excluded by a normal activated partial thromboplastin time or normal factor VIII or factor IX levels.535 A definitive molecular diagnosis combined with linkage analysis where necessary is needed, especially if prenatal or preimplantation diagnosis is sought. Determination of a pathogenic variant in the structurally complex factor VIII gene enables confirmation of carrier status.536, 537 Prenatal diagnosis requests for hemophilia A are uncommon, but have been provided.538–540 Preimplantation genetic testing (see Chapter 2) for hemophilia has also been accomplished.541 Noninvasive prenatal diagnosis of hemophilia A and B in hemophilia carriers using maternal plasma and factor VIII and factor IX sequence variants has been demonstrated542 (see Chapter 8). We all carry a host of deleterious recessive genes (∼100–300)543 and technical advances have enabled routine simultaneous testing of hundreds of autosomal recessive and X-linked disorders which affect about 1 in 300 pregnancies.544 Not well understood by patients is the fact that expanded carrier testing545–555 examines only a few common mutations in each gene analyzed. The net effect is a significant reduction in the risk of being a carrier of the gene tested. Unfortunately, the refrain heard from patients having had expanded carrier testing is “I am not a carrier.” Financial constraints prevent many couples benefitting from the extensive panel of carrier tests, leaving them with the previously required indications of ethnicity, affected offspring,

or family history. This type of limited carrier testing, which includes CF and spinal muscular atrophy, misses about 70 percent of carriers of rare disorders.556 For the most part carriers of autosomal recessive disorders are asymptomatic. An important exception are the carriers of the sickle cell disease gene mutation p.Glu6-Val in the β-globin chain of hemoglobin, who have an increased risk of both venous thromboembolism and chronic renal disease.557 This is an important realization that should lead to care and surveillance, given that about 300 million worldwide have the sickle cell trait. Autosomal recessive disease severity when due to compound heterozygous pathogenic variants will be a consequence of the variable expression of the two alleles (e.g. CF with the p.Phe508del and the p.Arg117His alleles resulting only in CBAVD) (see Chapter 15). Gene modifiers too will affect the phenotype. Variant interpretation remains a challenge as well as increasing the need and time taken for genetic counseling given that over 1,800 autosomal recessive genes are known.543 Clearly, the purpose of expanded carrier screening (see Chapter 14) for healthy couples enables them to benefit from available options that include preimplantation genetic testing, routine prenatal diagnosis, adoption, donor sperm or ova, or surrogacy. This approach has proved acceptable to the American College of Obstetricians and Gynecologists, the American College of Medical Genetics and Genomics, the Society for Maternal-Fetal Medicine, and the National Society of Genetic Counselors.558, 559 The clinical utility and efficacy has been clearly demonstrated.546, 549, 551, 558 Johansen Taber et al.560 reported on the actions and reproductive outcomes of 391 at-risk couples from a tested population of over 270,000 using a panel of 176 genetic disorders. Over 75 percent who had preconception testing, planned or acted to avoid having an affected progeny. More than 50 percent of at-risk couples terminated pregnancies. Relying on a survey study, the authors acknowledge, has limitations so far as memory, response bias, and selection (infertility problems) are concerned. In a smaller study, others549 demonstrated the clear superiority of expanded carrier screening compared with ethnicity-based testing, with over threefold detection. Punj et al. offered preconception next-generation sequencing to determine

34 Genetic Disorders and the Fetus

carriers and found 12/71 couples at risk.548 Eight were carriers of hemochromatosis. These authors analyzed 728 genes in 202 individuals, 78 percent being determined to have at least one positive carrier result. In this exploratory study, which used a 148 gene-panel rather than the ACMG actionable panel of 59 genes, 3.5 percent of participants had a medically actionable variant548 (see Chapter 14). Applying their analysis to the ACMG panel, 2.9 percent had an actionable variant. Ethnicity-based carrier testing (Table 1.5) remains the only option for large swaths of the world’s population. Selective Ashkenazi Jewish mutation carrier testing, for example, for disorders listed in Table 1.5 do provide valuable but limited information, leading to options noted above. A study of 6,805 Jewish patients (Ashkenazi, Sephardi, and Mizrahi) having expanded carrier screening showed that 64.6 percent were identified as a carrier of one or more of 96 disorders562 (Table 1.6). The authors noted that >80 percent of the reported variants would have been missed by standard Ashkenazi Jewish screening protocols. One in 16 couples were identified as joint carriers with a 25 percent risk of having an affected child. A novel, likely pathogenic variant was seen in about 2.5 percent of patients tested. A whole-exome sequencing study of 123,136 cases examined carrier rates in six ethnic groups, focusing on 415 genes associated with severe recessive disorders.563 These authors found that 32.6 percent (East Asian) and 62.9 percent (Ashkenazi Jewish) were variant carriers of at least one of the 415 genes. A pan-ethnic screen using these 415 genes would identify up to 2.52 percent of at-risk couples. However, the limitations of ethnic-based carrier testing were revealed by a genetic ancestry analysis of >93,000 individuals having expanded carrier testing using a 96-gene panel.565 Nine percent of those tested had an ancestry from a lineage inconsistent with self-reported ethnicity. Multiple published reports on preconception or prenatal expanded carrier screening using large but variable-sized gene panels overwhelmingly support this approach above ethnicity-based testing.545, 549, 566–572 Although not currently required in preconception carrier screening, testing for hereditary cancer risk should be considered. A personal or family history of cancer as well as ethnicity currently

serves as an indication for screening. Autosomal dominant disorders are otherwise not usually subject to screening. In a study of 26,906 individuals in the Healthy Nevada Project screened for BRCA-related breast and ovarian cancer, Lynch syndrome, and familial hypercholesterolemia, 1.33 percent were found to be carriers of pathogenic or likely pathogenic variants.573 Moreover 90 percent of carriers had not been identified previously, and only 25.2 percent had a relevant family history. These three disorders determined by screening (not family history) are not usually considered for prenatal diagnosis or preimplantation genetic testing. However, other autosomal dominant disorders with manifestations in childhood (e.g. multiple endocrine neoplasia type 2B, familial adenomatous polyposis, long QT syndrome, cardiomyopathy) do qualify for preconception, preimplantation, and prenatal testing. A study of 23,179 individuals with a family history of cancer had next-generation sequencing using a 30-gene panel.574 A total of 2,811 pathogenic variants were found in 2,698 individuals for an overall pathogenic frequency of 11.6 percent. For those of Ashkenazi Jewish descent three-quarters of the pathogenic variants in the BRCA1 and BRCA2 genes would have been missed if only the routine three common founder mutations were tested. Geneticists and genetic counselors will attest to the frequent challenges they encounter faced by their patients’ difficulty comprehending genetic test results, implications, and options. On the heels of the technologic advances in genetics have come commercialization in the form of direct-to-consumer (DTC) testing. Few patients are cognizant of the commercialization realities that include selling of their data, receiving misleading results, being faced with incorrect, false-positive or false-negative results, a lack of informed consent, confidentiality, and privacy.575–580 There is a wide spectrum of laws that govern genetic testing in most countries, with special reference to laboratory accreditation, staff certification, genetic counseling requirements, and informed consent. In one study of identical twins there was a lack of concordance between laboratories.581 In an illustrative case, the result provided was actionable, but no action was taken by the recipient of the DTC communication.582 Ethical breaches, including testing of children, further complicate DTC practices.583

CHAPTER 1

Genetic Counseling: Preconception, Prenatal, and Perinatal 35

Table 1.5 Genetic disorders in various ethnic groups. Ethnic group

Genetic disorder

Africans (black)

Sickle cell disease and other disorders of hemoglobin α- and β-thalassemia Glucose-6-phosphate dehydrogenase deficiency Benign familial leucopenia High blood pressure (in females)

Afrikaners (white South Africans)

Variegate porphyria Fanconi anemia

American Indians (of British Columbia)

Cleft lip or palate (or both)

Amish/Mennonites

Ellis–Van Creveld syndrome Pyruvate kinase deficiency Hemophilia B

Armenians

Familial Mediterranean fever

Ashkenazi Jews

A-β-lipoproteinemia Bloom syndrome Breast cancer Canavan disease Colon cancer Congenital adrenal hyperplasia Dysferlinopathy (limb girdle muscular dystrophy 2B) Dystonia musculorum deformans Factor XI (PTA) deficiency Familial dysautonomia Familial hyperinsulinism Fanconi anemia (type C) Galactosemia Gaucher disease (adult form) Iminoglycinuria Joubert syndrome Maple syrup urine disease Meckel syndrome Niemann–Pick disease Pentosuria Retinitis pigmentosa 590 Tay–Sachs disease Warsaw Breakage syndrome 561

Chinese

Thalassemia (𝛼) Glucose-6-phosphate dehydrogenase deficiency (Chinese type) Adult lactase deficiency

36 Genetic Disorders and the Fetus

Table 1.5 (Continued) Ethnic group

Genetic disorder

Eskimos

E1 pseudocholinesterase deficiency Congenital adrenal hyperplasia

Finns

Aspartylglucosaminuria Congenital nephrosis

French Canadians

Neural tube defects Tay–Sachs disease

Irish

Neural tube defects Phenylketonuria Schizophrenia

Italians (northern)

Fucosidosis

Japanese and Koreans

Acatalasia Dyschromatosis universalis hereditaria Oguchi disease

Maori (Polynesians)

Clubfoot

Mediterranean peoples (Italians,

Familial Mediterranean fever

Greeks, Sephardic Jews, Armenians,

Glucose-6-phosphate dehydrogenase deficiency

Turks, Spaniards, Cypriots)

(Mediterranean type) Glycogen storage disease (type III) Thalassemia (mainly β)

Norwegians

Cholestasis-lymphedema Phenylketonuria

Yugoslavs (of the Istrian Peninsula)

Schizophrenia

Table 1.6 Residual risk values for diseases in Ashkenazi Jewish populations. 100% Ashkenazi

Probability of

Jewish carrier Disease

frequency

Detectability

Residual

affected fetus if

risk

parents pos/nega

Gaucher disease

1 in 15

0.95

1 in 281

1 in 1,124

Cystic fibrosis

1 in 23

0.94

1 in 368

1 in 1,472

Tay–Sachs disease

1 in 27

0.98

1 in 1,301

1 in 5,204

Familial dysautonomia

1 in 31

>0.99

1 in 3,001

1 in 12,004

Canavan disease

1 in 55

>0.97

1 in 1,801

1 in 7,204

Glycogen storage disease type 1a

1 in 64

0.95

1 in 1,261

1 in 5,044

Hyperinsulinemic hypoglycemia

1 in 68

0.90

1 in 671

1 in 2,684

Mucolipidosis IV

1 in 89

0.95

1 in 1,761

1 in 7,044

Maple syrup urine disease

1 in 97

0.95

1 in 1,921

1 in 7,684

Fanconi anemia

1 in 100

0.99

1 in 9,901

1 in 39,604

Dihydrolipoamide dehydrogenase deficiency

1 in 107

>0.95

1 in 2,121

1 in 8,484

Niemann–Pick disease type A

1 in 115

0.97

1 in 3,801

1 in 15,204 1 in 9,524

Usher syndrome type 3

1 in 120

>0.95

1 in 2,381

Bloom syndrome

1 in 134

0.99

1 in 13,301

1 in 53,204

Usher syndrome type 1F

1 in 147

≥0.75

1 in 585

1 in 2,340

Nemaline myopathy

1 in 168

>0.95

1 in 3,341

1 in 13,364

a One

parent is positive and one parent is negative by carrier screening.

Source: Modified from Scott et al.564

CHAPTER 1

Genetic Counseling: Preconception, Prenatal, and Perinatal 37

Professional organizations, aware of all these issues, have discouraged the use of DTC genetic testing. Position statements have accordingly been issued by the American College of Obstetricians and Gynecologists,584 the American College of Medical Genetics and Genomics,585 the Joint Society of Obstetricians and Gynecologists, and the Canadian College of Medical Genetics.586 A range of laws exist in Europe, with France and Germany banning DTC genetic testing.587 Serious concern has been expressed about the ethical, legal, and regulatory challenges of DTC testing in Ireland588 and Europe.589

A family history of a genetic disorder The explicit naming of a specific genetic disorder when the family history is being discussed facilitates evaluation and any possible testing. Difficulties are introduced when neither family nor previous physicians have recognized a genetic disorder within the family, sometimes revealed by expanded carrier screening591 or whole-exome sequencing.592 Such a disorder may be common (e.g. factor V Leiden deficiency) but nevertheless unrecognized. Clinical clues would include individuals in the family with deep-vein thrombosis, sudden death possibly due to a pulmonary embolus, and yet other individuals with recurrent pregnancy loss.593 Venous thromboembolism is the third leading cause of cardiovascular death in the United States, and provides additional insights into the genetic basis of unprovoked pulmonary embolism. Using whole-exome sequencing in 393 affected individuals and 6,114 controls, Desch et al.594 identified four genes (PROS1, STAB2, PROC, SERPINC1) with pathogenic variants, expanding the need for genetic testing given the history of thromboembolism. For some families, individuals with quite different apparent clinical features may, in fact, have the same disorder. Seventeen cancers in different organs in family members may not be recognized as manifestations of the same common mutation. In hereditary nonpolyposis colon/rectal cancer, various family members may suffer from other cancers including the uterus, ovary, breast, stomach, small bowel, ureter, melanoma, or salivary glands. Analysis of the five culprit genes in the proband would enable detection of the mutation, which could then be assayed in other family members at

risk. In another example, there may be two or more deceased family members who died from “kidney failure,” and another one or two who died from a cerebral aneurysm or a sudden brain hemorrhage. Adult polycystic kidney disease (APKD) may be the diagnosis, which will require further investigation by both ultrasound and DNA analysis. Moreover, two different genes for APKD have been identified (about 85 percent of cases due to APKD1 and close to 15 percent due to APKD2),595 and a rare third locus is known. In yet other families, a history of hearing impairment/deafness in some members and sudden death in others may translate to the autosomal recessive Jervell and Lange–Nielsen syndrome.596 This disorder is characterized by severe congenital deafness, a long QT interval, and large T waves, together with a tendency for syncope and sudden death due to ventricular fibrillation. Given that a number of genetic cardiac conduction defects have been recognized, a history of an unexplained sudden death in a family should lead to a routine electrocardiogram at the first preconception visit and possibly mutation analysis of at least 15 long QT syndrome genes.597 Other disorders in which sudden death due to a conduction defect might have occurred, with or without a family history of cataract or muscle weakness, should raise the suspicion of myotonic muscular dystrophy (see Chapter 31). Rare named disorders in a pedigree should automatically raise the question of the need for genetic counseling. We have seen instances (e.g. pancreatitis) in which, in view of its frequency, the disorder was simply ascribed to alcohol or idiopathic categories. Hereditary pancreatitis, although rare, is an autosomal dominant disorder for which several genes are known.598 Awareness of the clinical manifestations in carrier females of X-linked disorders is important given health and risk implications (Table 1.7). The pattern of inheritance of an unnamed disorder may signal a specific monogenic form of disease. For example, unexplained intellectual disability on either side of the female partner’s family calls for fragile X DNA carrier testing. Moreover, unexpected segregation of a maternal premutation may have unpredicted consequences, including reversion of the triplet repeat number to the normal range.671 Genetic counseling may be valuable, more especially because the phenomena

38 Genetic Disorders and the Fetus

Table 1.7 Signs in females who are carriers of selected X-linked recessive disease pertinent to prenatal diagnosis. Selected disorders

Key feature(s) that may occur

Selected references

Aarskog–Scott syndrome allelic with

Widow’s peak or short stature

599

Achromatopsia

Decreased visual acuity and myopia

600

Adrenoleukodystrophy

Neurologic and adrenal dysfunction

601, 602

Alport syndrome

Microscopic hematuria and hearing impairment

603

Ameliogenesis imperfecta,

Mottled enamel vertically arranged

604

Arthrogryposis multiplex congenita

Club foot, contractures, hyperkyphosis

605

ATRX syndrome 𝛼-thalessemia/ID

Mild intellectual disability, hemoglobin H inclusions

599, 606

Tapered fingers, short, widely spaced, flexed toes,

607

XLMR 16

hypomaturation type

syndrome Borjeson–Forssman–Lehmann syndrome

mild mental retardation

Choroideremia a

Chorioretinal dystrophy

608

Chondrodysplasia punctata 1

Mild intellectual disability, possible bone defects

599

Chronic granulomatous disease

Cutaneous and mucocutaneous lesions

609–611

Cleft palate

Bifid uvula

612

Conductive deafness with stapes

Mild hearing loss

613

Mild high-pitch hearing loss

599

Dilated cardiomyopathy

Cardiac failure

614

Duchenne/Becker muscular

Pseudohypertrophy, muscle weakness,

615–618

and short stature

fixation Deafness X-linked 1 allelic with Charcot-Marie-Tooth 5

dystrophy

cardiomyopathy/conduction defects

Dyskeratosis congenita

Retinal pigmentation

619

Ectodermal dysplasia

Variable severity of skin, hair, nails, and teeth

599

Emery–Dreifuss muscular dystrophy

Cardiomyopathy/conduction defects

620–622

Fabry disease

Angiokeratomas, corneal dystrophy, "burning"

623, 624

hands and feet, rhabdomyolysis FG syndrome

Anterior displaced anus, facial dysmorphism

625

Fragile X syndrome

Mild-to-moderate intellectual disability, behavioral

626–628

aberrations, schizoaffective disorder, premature ovarian failure, fragile X tremor ataxia syndrome, women and men premutation carriers G6PD deficiency

Hemolytic crises, neonatal hyperbilirubinemia

629

Hemophilia A and B

Bleeding tendency

630

Hypohydrotic ectodermal dysplasia

Sparse hair, decreased sweating

631, 632

Ichthyosis

Ichthyosis

633

KDM5C gene disease

Intellectual disability

634

Lissencephaly and agenesis of the

Epilepsy with subcortical band heterotopia

599

Lenticular cataracts

635

MASA syndrome/SPG1

Mild intellectual disability, abducted thumbs

599

McLeod neuroacanthocytosis

Chorea, late-onset cognitive decline

636

Menkes disease

Patchy kinky hair, hypopigmentation

637, 638

Myopia

Mild myopia

639

Nance–Horan syndrome b

Posterior Y-sutural cataracts and dental anomalies

640

Norrie disease

Retinal malformations

641

Ocular albinism type 1

Retinal/fundal pigmentary changes

642

corpus callosum Lowe syndrome

syndrome

(see Chapter 16)

CHAPTER 1

Genetic Counseling: Preconception, Prenatal, and Perinatal 39

Table 1.7 (Continued) Selected disorders

Key feature(s) that may occur

Selected references

Oculofaciodigital syndrome (OFD1)

Facial dysmorphism, abnormal digits, and polycystic

599

allelic with

kidneys

Simson–Galabia–Beheld syndrome 2 and Joubert syndrome Oligodontia

Hypodontia

643

Opitz G/BBB syndrome

Hypertelorism

644

Opitz–Kaveggia syndrome

Mild intellectual disability, hypertelorism

599

Ornithine transcarbamylase

Hyperammonemia, psychiatric/neurologic

645, 646

deficiency

manifestations

Ovarian cancer

Ovarian cancer

647

Pelizaeus–Merzbacher

Possible mild spasticity

648

Retinoschisis

Peripheral retinal changes

649

Retinitis pigmentosa

Night blindness, concentric reduction of visual field,

650, 651

pigmentary fundal degeneration, extinction of electroretinogram, cone disruption, vision loss MECP2-duplication syndrome

Intellectual disability, neuropsychiatric features,

652

endocrine abnormalities 653, 654

Simpson–Golabi–Behmel syndrome

Extra lumbar/thoracic vertebrae, accessory nipples,

Spinal and bulbar muscular atrophy

Muscle weakness and cramps

655

Split-hand/split-foot anomaly

Mild split-hand/split-foot anomaly

656

Spondyloepiphyseal dysplasia, late

Arthritis

657

Slight hypoplasia of ulnar side of hand and mild

658

facial dysmorphism

onset Ulnar hypoplasia with lobster-claw deficiency of feet

syndactyly of toes

Wiskott–Aldrich syndrome a

Abnormal platelets and lymphocytes

659, 660

X-linked intellectual disability

Mostly intellectual disability (many genes),

661–663

occasional short stature, hypertension, psychiatric symptoms X-linked mental retardation

Short stature, hypertelorism

599, 664, 665

X-linked mental retardation

Cerebellar hypoplasia, distinctive facies

666, 667

X-linked myotubular myopathy

Weakness, respiratory problems

668

X-linked protoporphyria

Life-long photosensitivity; liver disease

669

X-linked retinitis pigmentosa

Retinal changes

670

(OPHN1)

a Uncertain. b May

be same disorder.

of pleiotropism (several different effects from a single gene) and heterogeneity (a specific effect from several genes) may confound interpretation in any of these families. History of a previous child with intellectual disability with a diagnosis deemed “idiopathic” or of unknown cause after chromosomal, fragile X and biochemical analyses, is no longer tenable without whole-exome sequencing672, 673 (see Chapter 14). Over 700 genes involved in intellectual disability of monogenic origin have been recognized.674, 675

In a meta-analysis of 3,350 individuals with neurodevelopmental disorders676–678 the diagnostic yield was 36 percent using whole-exome sequencing. More recently, whole-exome sequencing for patients sent for a chromosomal microarray yielded diagnoses in about 27 percent of intellectual disability cases.676

Consanguinity A wide swath of the world’s population have high rates of consanguinity (50–70 percent of births to

40 Genetic Disorders and the Fetus

consanguineous parents). This especially applies to India, Pakistan, Bangladesh, the Middle East, and Africa. Medical literature is replete with examples of rare severe autosomal recessive disease in these populations. Where family history does not reveal unknown or hidden consanguinity, purposeful or incidental, significant runs of homozygosity seen on a chromosomal microarray (see Chapter 13) frequently will. In those instances, recognition of a shared gene and its mutation within a shared region may unexpectedly lead to a rare diagnosis. Not as well known, perhaps, is that shared variant homozygosity markedly reduces the fertility rate of close consanguineous couples.679 Consanguineous couples face increased risks of having children with autosomal recessive disorders; the closer the relationship, the higher the risks. A study in the United Arab Emirates of 2,200 women ≥15 years of age (with a consanguinity rate of 25–70 percent) concluded that the occurrence of malignancies, congenital abnormalities, intellectual disability, and physical handicap was significantly higher in the offspring of consanguineous couples.680, 681 The pooled incidence of all genetic defects, regardless of the degree of consanguinity, was 5.8 percent, in contrast with a nonconsanguineous rate of 1.2 percent, similar to an earlier study.681, 682 A Jordanian study also noted significantly higher rates of infant mortality, stillbirths, and congenital malformations among the offspring of consanguineous couples.683 A Norwegian study of first-cousin Pakistani parents yielded a relative risk for birth defects of about twofold.684 In that study, 28 percent of all birth defects were attributed to consanguinity. An observational study of 5,776 Indian newborns noted a birth defect prevalence of 11.4 per 1,000 births with a consanguinity rate of 44.74 percent.685 A study from Saudi Arabia, where the consanguinity rate exceeds 50 percent, focused on whole-exome sequencing of 2,219 families who had or had lost an affected fetus or child. The study group was constituted by 1,653 individual samples, 127 twosomes, 370 trios, 58 quads, and 11 others.686 They resolved many cases by determining known causal recessive genes and their mutations, but also discovering multiple previously unknown pathogenic variants. In addition, they recognized some genes that also had a dominant rather than recessive mode of inheritance. Their

prenatal diagnostic detection rate was 46.2 percent (30/65 cases), 87 percent of which were autosomal recessive. Whole-exome sequencing following discovery of a fetal anomaly not resolved by karyotyping or chromosomal microarray may well provide a precise diagnosis. In a study of 102 anomalous fetuses, a definitive or probable diagnosis was made in 21 (20.6 percent).687 A similar small study of 19 families with fetal anomalies yielded candidate variants in 12 (63 percent).688 A systematic evidence-based review of exome and genome sequencing for congenital anomalies or intellectual disability on behalf of the ACMG concluded that a change in patient management was observed in nearly all studies, including an impact on reproductive outcomes.689 The occurrence of rare, unusual or unique syndromes invariably raises questions about potential consanguinity and common ancestral origins. Clinical geneticists will frequently be cautious in these situations, providing potential recurrence risks of 25 percent. Consanguineous couples may opt for the entire gamut of prenatal tests to diminish even their background risks, with special focus on their ethnic-specific risks.690 Abnormal or concerning prenatal ultrasound observations in pregnancies by consanguineous couples may prompt prenatal whole-exome sequencing.691

Environmental exposures that threaten fetal health Concerns about normal fetal development after exposure to medications, alcohol, illicit drugs, chemical, infectious or physical agents, and/or maternal illness are among the most common reasons for genetic counseling during pregnancy. Many of these anxieties and frequently real risks could be avoided through preconception care. Public health authorities, vested with the care of the underprivileged in particular, need to focus their scarce resources on preconception and prenatal care and on the necessary public education regarding infectious diseases, immunization, nutrition, and genetic disorders. In preconception planning, careful attention to broadly interpreted fetal “toxins” is necessary, and avoidance should be emphasized. Alcohol, smoking, illegal drug use, certain medications, and X-ray exposure require discussion. Estimates of the prevalence of the fetal alcohol spectrum disorder

CHAPTER 1

Genetic Counseling: Preconception, Prenatal, and Perinatal 41

approximate 2 per 1,000 livebirths692 in the United States but in certain regions and countries rates reach as high as 10 percent.693–695 There is a limited list of known and proven human drug teratogens696 (see Chapter 3). Maternal use of specific teratogenic medications,697 such as isotretinoin, may be missed, unless the physician expressly inquires about them. Preconception advice to avoid heat exposure in early pregnancy is appropriate. Our observations showed a 2.9 relative risk for having a child with a NTD in mothers who used a hot tub during the first 6 weeks of pregnancy.698 High fever in the very early weeks of pregnancy is a potential teratogen698 and should be avoided and treated promptly. Animal studies show that commonly used drugs enter the fetal brain.699 A report from the Spanish Collaborative Study of Congenital Malformations noted a 2.8-fold increased risk of Down syndrome in the offspring of women ≥35 years of age and who were taking oral contraceptives when they became pregnant.700 Identification of preconception options

The time to deal with unwanted risks is not during the second trimester of pregnancy, as is so often the case in practice. Preconception counseling will identify specific risks and attendant options, which include the following: • Knowledge of family history • Attention to maternal health (e.g. diabetes control,701, 702 confirm cardiac and vascular normality) • Decision not to have children (includes consideration of vasectomy or tubal ligation) • Adoption • In vitro fertilization • Gamete intrafallopian tube transfer or allied techniques • Artificial insemination by donor • Ovum donation (includes surrogacy) • Intracytoplasmic sperm injection • Carrier detection tests • Noninvasive prenatal screening by fetal DNA in the maternal circulation • Naternal serum 𝛼-fetoprotein screening for NTDs • Prenatal diagnosis (CVS, amniocentesis, cordocentesis, ultrasound, MRI) • Preimplantation genetic testing

Fetal treatment or surgery for selected disorders Folic acid supplementation in periconceptional period (see Chapter 10) • Noninvasive prenatal testing (aneuploidy; monogenic disorders) • Selective abortion • •

Genetic counseling as a prelude to prenatal diagnosis The assumption that noninvasive prenatal testing for common chromosomal abnormalities (see Chapter 7) is a screening and not a diagnostic test, is unfortunately common. Many women receiving a normal report opt to avoid an amniocentesis. The vast majority will be vindicated, but some will complete pregnancy with a child having a disorder that could have been diagnosed in early gestation. Physicians and counselors are advised to remind women of this limitation, given that about half of all chromosomal abnormalities will be missed by the noninvasive screen.703 Prospective parents should understand their specific indication for prenatal tests and the limitations of such studies. Frequently, one or both members of a couple fail to appreciate how focused the prenatal diagnostic study will be. Either or both may have the idea that all causes of intellectual disability or congenital defects will be detected or excluded. It is judicious for the physician to urge that both members of a couple come for the consultation before CVS or amniocentesis. Major advantages that flow from this arrangement include a clearer perception by the partner regarding risks and limitations, a more accurate insight into his family history, and an opportunity to detect an obvious (although unreported or undiagnosed) genetic disorder of importance (e.g. Treacher–Collins syndrome, facioscapulohumeral dystrophy or one of the orofacial–digital syndromes). Women making an appointment for genetic counseling should be informed about the importance of having their partner with them for the consultation, avoiding subsequent misunderstanding about risks, options, and limitations. Before prenatal genetic studies are performed, a couple should understand the inherent limitations both of the laboratory studies and, when relevant, of ultrasound. For detection of chromosomal

42 Genetic Disorders and the Fetus

disorders, they should be aware of potential maternal cell admixture and mosaicism (see Chapter 11). When faced with potential X-linked hydrocephalus, microcephaly, or other serious X-linked disorders, and the realization of less than 100 percent certainty of diagnosis, couples may elect fetal sex determination as the basis for their decision to keep or terminate a pregnancy at risk. For some, neither chromosomal microarrays, biochemical assays, nor DNA analyses will provide results with 100 percent certainty. The time taken to determine the fetal karyotype or other biochemical parameters should be understood before amniocentesis. The known anxiety of this period can be appreciably aggravated by a long, unexpected wait for a result. The need for a second amniocentesis is rarer nowadays but, in some circumstances, fetal blood sampling remains an additional option that may need discussion. Despite the very unlikely eventuality that no result may be obtained because of failed cell culture or contamination, this issue should be mentioned. The potential possibility for false-positive or false-negative results should be carefully discussed when applicable. Any quandary stemming from the results of prenatal studies is best shared immediately with the couple. The role of the physician in these situations is not to cushion unexpected blows or to protect couples from information that may be difficult to interpret. All information available should be communicated, including the inability to accurately interpret the observations made. This is especially so with the use of the chromosomal microarray (see Chapter 13) and whole-exome sequencing (see Chapter 14). Cautions are appropriate with special reference to VOUS (see Chapter 14), that require in addition, parental samples to determine inherited or de novo changes. Other key issues to be considered by the genetic counselor and discussed when appropriate with the consultand follow. Informed consent

Consent for minor procedures including amniocentesis and CVS has been a requirement for decades and needs no repetition. However, the advent of chromosomal microarrays (see Chapter 13) and whole-exome sequencing for prenatal diagnosis (see Chapter 14) requires additional explanations and caveats. Informed consent for

these two technologies is focused on the potential results, not sampling risks and procedures. The specific issues primarily involve the interpretation of results, their significance, the small possibility of uncertain findings, test limitations, and incidental results. Chromosomal microarray testing adds up to 6–10 percent to a prenatal diagnosis result (see Chapter 13) beyond the 8–10 percent for routine karyotyping, and whole-exome sequencing when done after the ultrasound discovery of fetal structural abnormality adds an additional 6.2–80 percent.691, 704–708 This absurd range reflects very small case series, varying indications, and the presence of single or multiple fetal abnormalities. A more likely detection range would be between 8.5 and 32 percent.707, 708 Prenatal diagnosis using whole-exome sequencing (see Chapter 14) is primarily focused on pregnancies in which fetal structural abnormality has been observed. A much less frequent indication would be a recent or late diagnosis of a parent with a likely monogenic disorder characterized by genetic heterogeneity. No matter the indication, the informed consent obtained incorporates and extends current practice for chromosomal microarray tests. The decision to offer whole-exome sequencing will almost inevitably come on the heels of the detection of fetal abnormality and in an atmosphere of tension and anxiety. Any center offering whole-exome sequencing will have, of necessity, established their informed consent procedure. The following list of pointers are likely to find common ground: 1. Pre- and postgenetic counseling by a geneticist or genetic counselor is a prerequisite, with strict adherence to ethical standards.709, 710 2. Both parents should be in attendance. 3. Explanations should use simple language, no jargon, and be in the language of the parents (with an interpreter, if needed). 4. The details of the fetal abnormality, effect on a child (pain; disability), a progressive disorder or not, and life expectancy. 5. The use of targeted sequencing, trios, and gene panels will need explanations, including the reason and need for prior or simultaneous chromosomal microarrays. 6. The time needed to obtain a result.

CHAPTER 1

Genetic Counseling: Preconception, Prenatal, and Perinatal 43

7. The likely detection rate and the limitations of whole-exome sequencing (e.g. repeat expansion disorder; mosaicism). 8. The occurrence of false positives, false negatives, or error. 9. The unexpected discovery of nonpaternity or consanguinity. 10. The detection of a variant of unknown significance.709 11. A “secondary finding”711–714 unrelated to the original purpose of the analysis. 12. The opportunity for the parents to opt out of receiving “secondary findings”715 which they should understand may have personal important health implications. 13. The choice to refuse testing. Presymptomatic or predictive testing

Presymptomatic or predictive testing is available for a rapidly increasing number of disorders, especially neuromuscular and neurodegenerative (see Chapter 14). Huntington disease is the prototype, and predictive testing using guidelines promulgated by the World Federation of Neurology,716–719 the International Huntington Association, and the European Huntington Disease Network719 are well established. Various programs report that a majority of patients are able to cope when it is found that they are affected,225–230, 720, 721 and, at least after a 1-year follow-up, potential benefit has been shown even in those found to be at increased risk.722 A European collaborative study evaluated 180 known carriers of the Huntington disease gene mutation and 271 noncarriers, all of whom received a predictive test result. Although the follow-up was only 3 years for about half the group, pregnancies followed in 28 percent of noncarriers and only 14 percent of carriers.723 Prenatal diagnosis was elected by about two-thirds of those who were carriers. Genetic counseling for Huntington disease when intermediate alleles with 27–35 CAG repeats are determined, pose significant challenges.724 Intermediate CAG repeats have been associated with behavioral, movement, and cognitive problems.725–727 The concern is the unpredictable likelihood of expansion which might account for 7 percent of new mutations.724 Providing counseling for those with low penetrance alleles (36–39 CAG repeats) is no less challenging. Repeats in

this range are estimated to occur randomly in the general population with a frequency of about 1 in 400.728 For patients with 36–39 repeats considering prenatal diagnosis, many factors will need to be addressed. These include all options discussed earlier and uncertainty, penetrance, anticipation, age of onset, and life expectancy. Experienced geneticists with an established program that includes predictive/presymptomatic testing for Huntington disease should preferably be consulted. As others earlier,729 we remain very concerned about the use of a test that can generate a “no hope” result. Even in sophisticated programs offering Huntington disease tests, fewer than expected at-risk individuals requested testing.730 A multicenter Canadian collaborative study evaluated the uptake, utilization, and outcome of 1,061 predictive tests, 15 prenatal tests, and 626 diagnostic tests from 1987 to 2000. The uptake for predictive testing was about 18 percent (range 12.5–20.7 percent).731 Of the 15 who had prenatal tests, 12 had an increased risk, which led to pregnancy termination in all but one.731 The motivations leading to the very difficult decision to have or not to have a predictive test are being recognized as extremely complex.732 In a Danish study before DNA tests were available, one in 20 individuals at risk for Huntington disease committed suicide, more than double the population rate,733 highlighting earlier reports of high suicide rates734 and emphasizing the erosive effects of uncertainty. However, a worldwide assessment of suicide rates, suicide attempts, or psychiatric hospitalizations after predictive testing did not confirm a high rate of suicide.735 In their worldwide questionnaire study sent to predictive testing centers, the authors noted that 44 individuals (0.97 percent) among 4,527 tested had five suicides, 21 suicide attempts, and 18 hospitalizations for psychiatric reasons. All those who committed suicide had signs of Huntington disease, while 11 (52.4 percent) of the 21 individuals who attempted suicide were symptomatic. Suicidal ideation or attempts remain a devastating reality for Huntington disease, especially given the psychopathology in those affected.736, 737 Depression, anxiety, and bipolar disorder are not infrequent. Suicidal behavior may be about 12 times that in the population at large, reaching an estimated 20 percent.738, 739 Others have written about the psychologic burden

44 Genetic Disorders and the Fetus

created by knowledge of a disabling fatal disease decades before its onset.740–742 Hayden743 warned that it is inappropriate to introduce a predictive test that “has the potential for catastrophic reactions” without a support program, including pretest and post-test counseling and specified standards for laboratory analyses. In one study, 40 percent of individuals tested for Huntington disease and who received DNA results required psychotherapy.744 A 5-year longitudinal study of psychologic distress after predictive testing for Huntington disease focused on 24 carriers and 33 tested noncarriers. Mean distress scores for both carriers and noncarriers were not significantly different but carriers had less positive feelings.745 A subgroup of tested persons were found to have long-lasting psychologic distress. An interview study of 20 who tested negative for Huntington disease revealed reactions that included obvious relief and gratitude, wishes to have (more) children, and life changes that included pursuit of a career and ending an unhappy relationship.746 Negative reactions included survivor guilt with sadness and depression or a feeling of pressure to do something extraordinary with their lives. Homozygotes for Huntington disease are rare747, 748 and reported in one out of 1,007 patients (0.1 percent). Counseling a patient homozygous for Huntington disease about the 100 percent probability of transmitting the disorder to each child is equivalent to providing a nonrequested predictive test,749 while failing to inform the patient of the risks would be regarded as the withholding of critical information. Pretest counseling in such cases would take into consideration a family history on both sides and therefore be able to anticipate the rare homozygous eventuality. On the other hand, an increasing number of examples already exist (see Chapter 14) in which presymptomatic testing is possible and important to either the patient or future offspring or both. Uptake has been high by individuals at risk, especially for various cancer syndromes.750 Use of DNA linkage or mutation analysis for ADPKD751, 752 may lead to the diagnosis of an unsuspected associated intracranial aneurysm in 8 percent of cases (or 16 percent in those with a family history of intracranial aneurysm or subarachnoid hemorrhage753 ) and preemptive surgery, with avoidance of a life-threatening sudden cerebral hemorrhage.

It is worth noting that a subgroup of families has features similar to Marfan syndrome and that haplo-insufficiency of the PKD1 gene influences the transforming growth factor-β (TGFβ) signaling pathway.754 In a study of 141 affected individuals, 11 percent decided against bearing children on the basis of the risk.755 These authors noted that only 4 percent of at-risk individuals between 18 and 40 years of age would seek elective abortion for an affected fetus. The importance of accurate presymptomatic tests for potential at-risk kidney donors has been emphasized.756 Organ donation by a sibling of an individual with ADPKD, later found to be affected, has occurred more than once. Since the PKD1 gene abuts the tuberous sclerosis (TSC2) gene, heterozygous deletions may lead to a contiguous gene-deletion syndrome.757 Individuals at 50 percent risk for familial polyposis coli (with inevitable malignancy for those with this mutated gene) who undergo at least annual colonoscopy could benefit from a massive reduction in risk (from 50 percent to 93,000 individuals undergoing expanded carrier screening reveals limitations of ethnicity-based medical guidelines. Genet Med 2020;22(10):1694. 566. Singer A, Sagi-Dain L. Impact of a national genetic carrier-screening program for reproductive purposes. Acta Obstet Gynecol Scand 2020;99:802. 567. Shi L, Webb BD, Birch AH, et al. Comprehensive population screening in the Ashkenazi Jewish population for recurrent disease-causing variants. Clin Genet 2017;91:599. 568. Baskovich B, Hiraki S, Upadhyay K, et al. Expanded genetic screening panel for the Ashkenazi Jewish population. Genet Med 2016;18:522. 569. Bristow SL, Morris JM, Peyser A, et al. Choosing an expanded carrier screening panel: comparing two panels at a single fertility centre. Reprod Biomed Online 2019;38:225. 570. Rosenblum LS, Zhu H, Zhou Z, et al. Comparison of pan-ethnic and ethnic-based carrier screening panels for individuals of Ashkenazi Jewish descent. J Genet Couns 2020;29:56. 571. Arjunan A, Litwack K, Collins N, et al. Carrier screening in the era of expanding genetic technology. Genet Med 2016;18:1214.

572. Gregg AR, Edwards JG. Prenatal genetic carrier screening in the genomic age. Semin Perinatol 2018;42:303. 573. Grzymski JJ, Elhanan G, Morales Rosado JA, et al. Population genetic screening efficiently identifies carriers of autosomal dominant diseases. Nat Med 2020;26(8):1235. 574. Neben CL, Zimmer AD, Stedden W, et al. Multi-gene panel testing of 23,179 individuals for hereditary cancer risk identifies pathogenic variant carriers missed by current genetic testing guidelines. J Mol Diagn 2019;21:646. 575. Tandy-Connor S, Guiltinan J, Krempely K, et al. False-positive results released by direct-to-consumer genetic tests highlight the importance of clinical confirmation testing for appropriate patient care. Genet Med 2018;20:1515. 576. Petersen LM, Lefferts JA. Lessons learned from direct-to-consumer genetic testing. Clin Lab Med 2020;40:83. 577. Laestadius LI, Rich JR, Auer PL. All of your data (effectively) belongs to us: data practices among direct-to-consumer genetic testing firms. Genet Med 2017;19:513. 578. de Pauw A, Schwartz M, Colas C, et al. Direct-to-consumer misleading information on cancer risks calls for an urgent clarification of health genetic testing performed by commercial companies. Eur J Cancer 2020;132:100. 579. Horton R, Crawford G, Freeman L, et al. Direct-to-consumer genetic testing. BMJ 2019;367:I5688. 580. Millward M, Tiller J, Bogwitz M, et al. Impact of direct-to-consumer genetic testing on Australian clinical genetic services. Eur J Med Genet 2020;63:103968. 581. Huml AM, Sullivan C, Figueroa M, et al. Consistency of direct-to-consumer genetic testing results among identical twins. Am J Med 2020;133:143.e2. 582. Garmany R, Lee CJ, Sharp RR, et al. Failure to follow up on a medically actionable finding from direct to consumer genetic testing: a case report. Mol Genet Genomic Med 2020;8:e.1252. 583. Niemiec E, Kalokairinou L, Howard HC. Current ethical and legal issues in health-related direct-to-consumer genetic testing. Per Med 2017;14:433. 584. Committee on Genetics. Committee opinion no. 724: consumer testing for disease risk. Obstet Gynecol 2017;130:e270. 585. ACMG Board of Directors. Direct-to-consumer genetic testing: a revised position statement of

CHAPTER 1

586.

587.

588. 589.

590.

591. 592.

593.

594.

595.

596.

597.

598.

599.

Genetic Counseling: Preconception, Prenatal, and Perinatal 85

the American College of Medical Genetics and Genomics. Genet Med 2016;18:207. Wilson RD, De Bie I, Armour CM, et al. Joint SOGC-CCMG opinion for reproductive genetic carrier screening: an update for all Canadian providers of maternity and reproductive healthcare in the era of direct-to-consumer testing. J Obstet Gynaecol Can 2016;38:742.e3. Kalokairinou L, Howard HC, Slokenberga S, et al. Legislation of direct-to-consumer genetic testing in Europe: a fragmented regulatory landscape. J Community Genet 2018;9:117. de Paor A. Direct to consumer genetic testing-law and policy concerns in Ireland. Ir J Med Sci 2018;187:575. Rafiq M, Ianuale C, Ricciardi W, et al. Direct-to-consumer genetic testing: a systematic review of European guidelines, recommendations, and position statements. Genet Test Mol Biomarkers 2015;19:535. Tatour Y, Tamaiev J, Shamaly S, et al. A novel intronic mutation of PDE6B is a major cause of autosomal recessive retinitis pigmentosa among Caucasus Jews. Mol Vis 2019;25:155. Sparks TN. Expanded carrier screening: counseling and considerations. Hum Genet 2020;139:1131. Tayoun AA, Mason-Suares H. Considerations for whole exome sequencing unique to prenatal care. Hum Genet 2020;139:1149. Simioni P, Prandoni P, Lensing AW, et al. The risk of recurrent venous thromboembolism in patients with an Arg5066Glnmutation in the gene for factor V (factor V Leiden). N Engl J Med 1997;336:399. Desch KC, Ozel AB, Halvorsen M, et al. Whole-exome sequencing identifies rare variants in STAB2 associated with venous thromboembolic disease. Blood 2020;136:533. Veldhuisen B, Saris JJ, deHaij S, et al. A spectrum of mutations in the second gene for autosomal dominant polycystic kidney disease (PKD2). Am J Hum Genet 1997;61:547. Neyroud N, Tesson F, Denjoy I, et al. A novel mutation in the potassium channel gene KVLQT1 causes the Jervell and Lange–Nielsen cardioauditory syndrome. Nat Genet 1997;15:186. Wallace E, Howard L, Liu M, et al. Long QT syndrome: genetics and future perspective. Pediatr Cardiol 2019;40:1419. Rebours V, Boutron-Ruault MC, Schnee M, et al. The natural history of hereditary pancreatitis: a national series. Gut 2009;58:97. Migeon BR. X-linked diseases: susceptible females. Genet Med 2020;22:1156.

600. Pinckers A. X-linked progressive cone dystrophy. Doc Ophthalmol Proc Ser 1982;33:399. 601. El-Deiry SS, Naidu S, Blevins LS, et al. Assessment of adrenal function in women heterozygous for adrenoleukodystrophy. J Clin Endocrinol Metab 1997;82:856. 602. Menage P, Carreau V, Tourbah A, et al. Les adrenoleucodystrophies heterozygotes symptomatiques de l’adulte: 10 cas. Rev Neurol 1993;149:445. 603. Hasstedt SJ, Atkin CL, San Juan AC Jr., Genetic heterogeneity among kindreds with Alport syndrome. Am J Hum Genet 1986;38:940. 604. Patel RR, Hovijitra S, Kafrawy AH, et al. X-linked (recessive) hypomaturation amelogenesis imperfecta: a prosthodontic, genetic and histopathologic report. J Prosthet Dent 1991;66:398. 605. Hennekam RC, Barth PG, van Lookeren Campagne W, et al. A family with severe X-linked arthrogryposis. Eur J Pediatr 1991;150:656. 606. Gibbons RJ, Suthers GK, Wilkie AOM, et al. X-linked alpha-thalassemia/mental retardation (ATR-X) syndrome: location to Xq12–q21.31 by X inactivation and linkage analysis. Am J Hum Genet 1992;51:1136. 607. Mathews KD, Ardinger HH, Nishimura DY, et al. Linkage localization of Borjeson–Forssman–Lehmann syndrome. Am J Med Genet 1989;34:470. 608. Karna J. Choroideremia: a clinical and genetic study of 84 Finnish patients and 126 female carriers. Acta Ophthalmol 1986;176(Suppl):1. 609. Romera MG, Martin MM, Gonzalez E. Chronic granulomatous disease: a case study of a symptomatic carrier. J Invest Allerg Clin Immunol 1997;7:57. 610. Lovas JG, Issekutz A, Walsh N, et al. Lupus erythematosus-like oral mucosal and skin lesions in a carrier of chronic granulomatous disease. Chronic granulomatous disease carrier genodermatosis. Oral Surg Oral Med Oral Pathol Oral Rad Endod 1995;80:78. 611. Marciano BE, Zerbe CS, Falcone EL, et al. X-linked carriers of chronic granulomatous disease: illness, lyonization, and stability. J Allergy Clin Immunol 2018;141:365. 612. Rollnick BR, Kaye CI. Mendelian inheritance of isolated nonsyndromic cleft palate. Am J Med Genet 1986;24:465. 613. Cremers CWRJ, Huygen PLM. Clinical features of female heterozygotes in the X-linked mixed deafness syndrome (with perilymphatic gusher during stapes surgery). Int J Pediatr Otorhinolaryngol 1983;6:179. 614. Kamdar F, Garry DJ. Dystrophin-deficient cardiomyopathy. J Am Coll Cardiol 2016;67:2533.

86 Genetic Disorders and the Fetus

615. Politano L, Nigro V, Nigro G, et al. Development of cardiomyopathy in female carriers of Duchenne and Becker muscular dystrophies. JAMA 1996;275:1335. 616. Matthews PM, Benjamin D, van Bakel I, et al. Muscle X-inactivation patterns and dystrophin expression in Duchenne muscular dystrophy carriers. Neuromuscul Disord 1995;5:209. 617. Azofeifa J, Voit T, Hubner C, et al. X-chromosome methylation in manifesting and healthy carriers of dystrophinopathies: concordance of activation ratios among first degree female relatives and skewed inactivation as cause of the affected phenotypes. Hum Genet 1995;96;167. 618. Darras BT, Urion DK, Ghosh PS. Dystrophinopathies. In: Adam MP, Ardinger HH, Pagon RA, et al., eds. GeneReviews [internet]. Seattle (WA): University of Washington, Seattle, 1993–2020, September 5, 2000. 619. Schnur RE, Heymann WR. Reticulate hyperpigmentation. Semin Cutan Med Surg 1997;16:72. 620. Emery AEH. Emery–Dreifuss syndrome. J Med Genet 1989;26:637. 621. Viggiano E, Madej-Pilarczyk A, Carboni N, et al. X-linked Emery-Dreifuss muscular dystrophy: study of X-chromosome inactivation and its relation with clinical phenotypes in female carriers. Genes (Basel) 2019;10:919. 622. Kong D, Zhan Y, Liu C, et al. A novel mutation of the EMD gene in a family with cardiac conduction abnormalities and a high incidence of sudden cardiac death. Pharmgenomics Pers Med 2019;12:319. 623. Ropers HH, Wienker TF, Grimm T, et al. Evidence for preferential X-chromosome inactivation in a family with Fabry disease. Am J Hum Genet 1977;29:361. 624. Berger JR. Misdiagnosis of multiple sclerosis in a female heterozygote with Fabry’s disease. Mult Scler Relat Disord 2019;30:45. 625. Thompson EM, Baraitser M, Lindenbaum RH, et al. The FG syndrome: seven new cases. Clin Genet 1985;27:582. 626. Vianna-Morgante AM, Costa SS, Pares AS, et al. FRAXA premutation associated with premature ovarian failure. Am J Med Genet 1996;64:373. 627. Sobesky WE, Taylor AK, Pennington BF, et al. Molecular/clinical correlations in females with fragile X. Am J Med Genet 1996;64:340. 628. Franke P, Maier W, Hautzinger M, et al. Fragile-X carrier females: evidence for a distinct psychopathological phenotype? Am J Med Genet 1996;64:334. 629. Meloni T, Forteleoni G, Dore A, et al. Neonatal hyperbilirubinaemia in heterozygous glucose-6-phosphate

630.

631. 632.

633.

®

634.

635. 636.

637.

638.

639. 640.

641.

642.

643.

644.

dehydrogenase deficient females. Br J Haematol 1983;53:241. Mauser Bunchoten EP, van Houwelingen JC, Sjamsoedin Visser EJ, et al. Bleeding symptoms in carriers of hemophilia A and B. Thromb Haemost 1988;59:349. Vabres P, Larregu M. X-linked genodermatoses. Ann Dermatol Venereol 1995;122:154. Rahbaran M, Doabsari MH, Salavitabar S, et al. A novel frameshift mutation in the EDA gene in an Iranian patient affected by X-linked hypohidrotic ectodermal dysplasia. Call Mol Biol Lett 2019;24:54. Afzal S, Ramzan K, Ullah S, et al. A novel nonsense mutation in the STS gene in a Pakistani family with X-linked recessive ichthyosis: including a very rare case of two homozygous female patients. BMC Med Genet 2020;21:20. Carmignac V, Nambot S, Lehalle D, et al. Further delineation of the female phenotype with KDM5C disease causing variants: 19 new individuals and review of the literature. Clin Genet 2020;98:43. Endres W. Inherited metabolic diseases affecting the carrier. J Inherit Metab Dis 1997;20:9. Jung HH, Danek A, Walker RH, et al. McLeod neuroacanthocytosis syndrome. GeneReviews . Seattle (WA): University of Washington, Seattle, 1993–2020, 2004 [updated 2019 May 23]. Lorette G, Toutain A, Barthes M, et al. Menkes syndrome: an unusual pigmentation anomaly in a mother and three sisters. Ann Pediatr 1992;39:453. Collie WR, Moore CM, Goka TJ, et al. Pili torti as marker for carriers of Menkes disease. Lancet 1978;1:607. Bartsocas CS, Kastrantas AD. X-linked form of myopia. Hum Hered 1981;31:199. Nance WE, Warburg M, Bixler D, et al. Congenital X-linked cataract, dental anomalies and brachymetacarpalia. Birth Defects Orig Artic Ser 1974;10:285. Kellner U, Fuchs S, Bornfeld N, et al. Ocular phenotypes associated with two mutations (R121W, C126X) in the Norrie disease gene. Ophthalmol Genet 1996;17:67. Charles SJ, Moore AT, Zhang Y, et al. Carrier detection in X-linked ocular albinism using linked DNA polymorphisms. Br J Ophthalmol 1994;78:539. Erpenstein H, Pfeiffer RA. Geschlechsgebunden-dominant erbliche Zahnunterzahl. Humangenetik 1967;4:280. Meroni G. X-linked Opitz G/BBB syndrome. GeneReviews Seattle (WA): University of

®

®

CHAPTER 1

645.

646.

647.

648.

649.

650.

651.

652.

653.

654.

Washington, Seattle, 1993–2020, 2004 [updated 2018 Apr 5]. Herinklake S, Boker K, Manns M. Fatal clinical course of ornithine transcarbamylase deficiency in an adult heterozygous female patient. Digestion 1977;58:83. Fries MH, Kuller JA, Jurecki E, et al. Prenatal counseling in heterozygotes for ornithine transcarbamylase deficiency in an adult heterozygous female patient. Digestion 1997;58:83. Etter JL, Moysich K, Kohli S, et al. Transmission of X-linked ovarian cancer: characterization and implications. Diagnostics (Basel) 2020;10:90. Warshawsky I, Rudick RA, Staugaitis SM, et al. Primary progressive multiple sclerosis as a phenotype of a PLP1 gene mutation. Ann Neurol 2005;58:470. Kaplan J, Pelet A, Hentari H, et al. Contribution to carrier detection and genetic counseling in X linked retinoschisis. J Med Genet 1991;28:383. Souied E, Segues B, Ghazi I, et al. Severe manifestations in carrier females in X linked retinitis pigmentosa. J Med Genet 1997;34:793. Kurata K, Hosono K, Hayashi T, et al. X-linked retinitis pigmentosa in Japan: clinical and genetic findings in male patients and female carriers. Int J Mol Sci 2019;20:1518. Ramocki MB, Peters SU, Tavyev YJ, et al. Autism and other neuropsychiatric symptoms are prevalent in individuals with MeCP2 duplication syndrome. Ann Neurol 2009;66:771. McKusick VA. Mendelian inheritance in man, 11th edn. Baltimore, MD: Johns Hopkins University Press, 1994:2530. Sajorda BJ, Gonzalez-Gandolfi CX, Hathaway ER, et al. Simpson-Golabi-Behmel syndrome type 1. GeneReviews Seattle (WA): University of Washington, Seattle, 1993–2020, 2006 [updated 2018 Nov 29]. Ishihara H, Kanda F, Nishio H, et al. Clinical features and skewed X-chromosome inactivation in female carriers of X-linked recessive spinal and bulbar muscular atrophy. J Neurol 2001;248:856. Ahmad M, Abbas H, Haque S, et al. X-chromosomally inherited split-hand/split foot anomaly in a Pakistani kindred. Hum Genet 1987;75:169. McKusick VA. Mendelian inheritance in man, 11th edn. Baltimore, MD: Johns Hopkins University Press, 1994:2535. van den Berghe H, Dequeker J, Fryns JP, et al. Familial occurrence of severe ulnar aplasia and lobster claw feet: a new syndrome. Hum Genet 1978;42:109. Wengler G, Gorlin JB, Williamson JM, et al. Nonrandom inactivation of the X chromosome in

®

655.

656.

657.

658.

659.

Genetic Counseling: Preconception, Prenatal, and Perinatal 87

early lineage hematopoietic cells in carriers of Wiskott–Aldrich syndrome. Blood 1995;85:2471. 660. Peacocke M, Siminovitch KA. The Wiskott–Aldrich syndrome. Semin Dermatol 1993;12:247. 661. Ziats CA, Schwartz CE, Gecz J, et al. X-linked intellectual disability: phenotypic expression in carrier females. Clin Genet 2019;97:418. 662. Uliana V, Bonatti F, Zanatta V, et al. Spectrum of X-linked intellectual disabilities and psychiatric symptoms in a family harbouring a Xp22.12 microduplication encompassing the RPS6KA3 gene. J Genet 2019;98:10. 663. Jackson MR, Loring KE, Homan CC, et al. Heterozygous loss of function of IQSEC2/ Iqsec2 leads to increased activated Arf6 and severe neurocognitive seizure phenotype in females. Life Sci Alliance 2019;2:e.201900386. 664. Stoll C, Geraudel A, Chauvin A. New X-linked syndrome of mental retardation, short stature and hypertelorism. Am J Med Genet 1991;39:474. 665. Atkin JF, Flaitz K, Patil S, et al. A new X-linked mental retardation syndrome. Am J Med Genet 1985;21:697. 666. Barresi S, Tomaselli S, Athanasiadis A, et al. Oligophrenin-1 (OPHN1), a gene involved in X-linked intellectual disability, undergoes RNA editing and alternative splicing during human brain development. PLoS One 2014;9:e91351. 667. Al-Owain M, Kaya N, Al-Zaidan H, et al. Novel intragenic deletion in OPHN1 in a family causing XLMR with cerebellar hypoplasia and distinctive facial appearance. Clin Genet 2011;79:363. 668. Dowling JJ, Lawlor MW, Das S. X-linked myotubular

®

myopathy. GeneReviews

Seattle (WA): University

of Washington, Seattle, 1993–2020, 2002 [updated 2018 Aug 23]. 669. Brancaleoni V, Balwani M, Granata F, et al. X-chromosomal inactivation directly influences the phenotypic manifestation of X-linked protoporphyria. Clin Genet 2016;89:20. 670. Nowakowski R. Ocular manifestations in female carriers of X-linked disorders. J Am Optom Assoc 1995;66:352. 671. Mornet E, Chateau C, Taillandier A, et al. Recurrent and unexpected segregation of the FMR1 CGG repeat in a family with fragile X syndrome. Hum Genet 1996;97:512. 672. Bowling KM, Thompson ML, Amaral MD, et al. Genomic diagnosis for children with intellectual disability and/or developmental delay. Genome Med 2017;9:43.

88 Genetic Disorders and the Fetus

673. Sun Y, Ruivenkamp CAL, Hoffer MJV, et al. Next-generation diagnostics: gene panel, exome, or whole genome? Hum Mutat 2015;36:648. 674. Vissers LELM, Gilissen C, Veltman JA. Genetic studies in intellectual disability and related disorders. Nat Rev Genet 2016;17:9. 675. Mir YR, Kuchay RAH. Advances in identification of genes involved in autosomal recessive intellectual disability: a brief review. J Med Genet 2019;56:567. 676. Linstrand A, Eisfeldt J, Pettersson M, et al. From cytogenetics to cytogenomics: whole-genome sequencing as a first-line test comprehensively captures the diverse spectrum of disease-causing genetic variation underlying intellectual disability. Genome Med 2019;11:68. 677. Miller DT, Adam MP, Aradhya S, et al. Consensus statement: chromosomal microarray is a first-tier clinical diagnostic test for individuals with developmental disabilities or congenital anomalies. Am J Hum Genet 2010;86(5):749. 678. Waggoner D, Wain KE, Dubuc AM, et al. Yield of additional genetic testing after chromosomal microarray for diagnosis of neurodevelopmental disability and congenital anomalies: a clinical practice resource of the American College of Medical Genetics and Genomics (ACMG). Genet Med 2018;20(10):1105. 679. Clark DW, Okada Y, Moore KHS, et al. Associations of autozygosity with a broad range of human phenotypes. Nat Commun 2019;10:4957. 680. Abdulrazzaq YM, Bener A, Al-Gazali LI, et al. A study of possible deleterious effects of consanguinity. Clin Genet 1997;51:167. 681. Saadallah AA, Rashed MS. Newborn screening: experiences in the Middle East and North Africa. J Inherit Metab Dis 2007;30:482. 682. Bundey S, Alam H, Kaur A, et al. Race, consanguinity and social features in Birmingham babies: a basis for prospective study. J Epidemiol Commun Health 1990;44:130. 683. Khoury SA, Massad DF. Consanguinity, fertility, reproductive wastage, infant mortality and congenital malformations in Jordan. Saudi Med J 2000;21:150. 684. Stoltenberg C, Magnus P, Lie RT, et al. Birth defects and parental consanguinity in Norway. AmJ Epidemiol 1997;145:439. 685. Perveen F, Tyyab S. Frequency and pattern of distribution of congenital anomalies in the newborn and associated maternal risk factors. J Coll Physicians Surg Pak 2007;17:340. 686. Monies D, Abouelhoda M, Assoum M, et al. Lessons learned from large-scale, first-tier clinical exome

687.

688.

689.

690. 691.

692.

693.

694.

695.

696. 697.

698.

699.

700.

701.

sequencing in a highly consanguineous population. Am J Hum Genet 2019;104(6):1182. Vora NL, Gilmore K, Brandt A, et al. An approach to integrating exome sequencing for fetal structural anomalies into clinical practice. Genet Med 2020;22(5):954. Meier N, Bruder E, Lapaire O, et al. Exome sequencing of fetal anomaly syndromes: novel phenotype-genotype discoveries. Eur J Hum Genet 2019;27:730. Malinowski J, Miller DT, Demmer L, et al. Systematic evidence-based review: outcomes from exome and genome sequencing for pediatric patients with congenital anomalies or intellectual disabilities. Genet Med 2020;22(6):986. Milunsky A. Heredity and your family’s health. Baltimore, MD: Johns Hopkins University Press, 1992. Greenbaum L, Pode-Shakked B, Eisenberg-Barzilai S, et al. Evaluation of diagnostic yield in fetal whole-exome sequencing: a report on 45 consecutive families. Front Genet 2019;10:425. Goh YI, Chudley AE, Clarren SK, et al. Development of Canadian screening tools for fetal alcohol spectrum disorder. Can J Clin Pharmacol 2008;15:e344. Memo L, Gnoato E, Caminiti S, et al. Fetal alcohol spectrum disorders and fetal alcohol syndrome: the state of the art and new diagnostic tools. Early Hum Dev 2013;89(Suppl 1):S40. Chudley AE. Fetal alcohol spectrum disorder: counting the invisible – mission impossible? Arch Dis Child 2008;93:721. Clarren SK, Randels SP, Sanderson M, et al. Screening for fetal alcohol syndrome in primary schools: a feasibility study. Teratology 2001;63:3. Holmes LB. Human teratogens: update 2010. Birth Defects Red A Clin Mol Teratol 2011;91:1. Gheysen W, Kennedy D. An update on maternal medication-related embryopathies. Prenat Diagn 2020;40:1168. Milunsky A, Ulcickas M, Rothman KJ, et al. Maternal heat exposure and neural tube defects. JAMA 1992;268:882. Saunders NR, Dziegielewska KM. Medications for pregnant women: A balancing act between the interests of the mother and of the fetus. Prenat Diagn 2020;40:1156. Martinez-Frias ML, Bermejo E, Rodriguez-Pinilla E, et al. Periconceptional exposure to contraceptive pills and risk for Down syndrome. J Perinatol 2001;21:288. Shub A, Lappas M. Pregestational diabetes in pregnancy: complications, management, surveillance, and

CHAPTER 1

702.

703.

704.

705.

706.

707.

708.

709.

710.

711.

712.

713.

714.

Genetic Counseling: Preconception, Prenatal, and Perinatal 89

mechanisms of disease – a review. Prenat Diagn 2020;40:1092. Murray SR, Reynolds RM. Short- and long-term outcomes of gestational diabetes and its treatment on fetal development. Prenat Diagn 2020;40:1085. Evans MI, Andriole S, Curtis J, et al. The epidemic of abnormal copy number variant cases missed because of reliance upon noninvasive prenatal screening. Prenat Diagn 2018;38(10):730. Vora NL, Powell B, Brandt A, et al. Prenatal exome sequencing in anomalous fetuses: new opportunities and challenges. Genet Med 2017;19:1207. Jelin AC, Vora N. Whole exome sequencing: applications in prenatal genetics. Obstet Gynecol Clin North Am 2018;45:69. Best S, Wou K, Vora N, et al. Promises, pitfalls and practicalities of prenatal whole exome sequencing. Prenat Diagn 2018;38:10. Normand EA, Braxton A, Nassef S, et al. Clinical exome sequencing for fetuses with ultrasound abnormalities and suspected Mendelian disorder 2018;10:74. Lord J, McMullan DJ, Eberhardt RY, et al. Prenatal exome sequencing analysis in fetal structural anomalies detected by ultrasonography (PAGE): a cohort study. Lancet 2019;393:747. Horn R, Parker M. Opening Pandora’s box?: ethical issues in prenatal whole genome and exome sequencing. Prenat Diagn 2018;38:20. Bunnik EM, de Jong A, Nijsingh N, et al. The new genetics and informed consent: differentiating choice to preserve autonomy. Bioethics 2013;27:348. ACMG Board of Directors. Points to consider in the clinical application of genomic sequencing. Genet Med 2012;14:759. Committee on Genetics and the Society for Maternal-Fetal Medicine. Committee opinion no. 682: microarrays and next-generation sequencing technology: the use of advanced genetic diagnostic tools in obstetrics and gynecology. Obstet Gynecol 2016;128:e262. Kalia SS, Adelman K, Bale SJ, et al. Recommendations for reporting of secondary findings in clinical exome and genome sequencing, 2016 update (ACMG SF v2.0): a policy statement of the American College of Medical Genetics and Genomics. Genet Med 2017;19:249. Johnston JJ, Rubinstein WS, Facio FM, et al. Secondary variants in individuals undergoing exome sequencing: screening of 572 individuals identifies high-penetrance mutations in cancer-susceptibility genes. Am J Hum Genet 2012;91:97.

715. Pinxten W, Howard HC. Ethical issues raised by whole genome sequencing. Best Pract Res Clin Gastroenterol 2014;28:269. 716. Committee of the International Huntington Association and the World Federation of Neurology. Guidelines for the molecular genetics predictive test in Huntington’s disease. J Med Genet 1994;31:555. 717. International Huntington Association and World Federation of Neurology. Guidelines for the molecular genetics predictive test in Huntington’s disease. Neurology 1994;44:1533. 718. Skirton H, Goldsmith L, Jackson L, et al. Quality in genetic counseling for presymptomatic testing-clinical guidelines for practice across the range of genetic conditions. Eur J Hum Genet 2013;21:256. 719. MacLeod RTA, Frontali M, Evers-Kiebooms G, et al. Recommendations for the predictive genetic test in Huntington’s disease. Clin Genet 2013;23:221. 720. Tibben A, Duivenvoorden J, Niermeijer MF, et al. Psychological effects of presymptomatic DNA testing for Huntington’s disease in theDutch program. Psychosom Med 1994;56:526. 721. Lawson K, Wiggins S, Green T, et al. Adverse psychological events occurring in the first year after predictive testing for Huntington’s disease. J Med Genet 1996;33:856. 722. Wiggins S, Whyte P, Huggins M, et al. The psychological consequences of predictive testing for Huntington’s disease. N Engl J Med 1992;327:1401. 723. Evers-Kiebooms G, Nys K, Harper P, et al. Predictive DNA-testing for Huntington’s disease and reproductive decision making: a European collaborative study. Eur J Hum Genet 2002;10:167. 724. Migliore S, Jankovic J, Squitieri F. Genetic counseling in Huntington’s disease: potential new challenges on horizon? Front Neurol 2019;10:453. 725. Kenney C, Powell S, Jankovic J. Autopsy-proven Huntington’s disease with 29 trinucleotide repeats. Mov Dis 2007;22:127. 726. Killoran A, Biglan KM, Jankovic J, et al. Characterization of the Huntington intermediate CAG repeat expansion phenotype in PHAROS. Neurology 2013;80:2022. 727. Cubo E, Ramos-Arroyo MA, Martinez-Horta S, et al. Intermediate CAG repeats in Huntington’ s disease. A longitudinal analysis of the European Huntington’ s Disease Network Registry Cohort (S25. 003). Neurology 2016;86:571. 728. Kay C, Collins JA, Miedzybrodzka Z, et al. Huntington disease reduced penetrance alleles occur at

90 Genetic Disorders and the Fetus

729. 730.

731.

732.

733.

734.

735.

736.

737.

738.

739.

740.

741.

742.

high frequency in the general population. Neurology 2016;87:282. Perry TL. Some ethical problems in Huntington’s chorea. Can Med Assoc J 1981;125:1098. Quaid KA, Brandt J, Faden RR, et al. Knowledge, attitude and the decision to be tested for Huntington’s disease. Clin Genet 1989;36:431. Moore RC, Xiang F, Monaghan J, et al. Huntington disease phenocopy is a familial prion disease. Am J Hum Genet 2001;69:1385. Evers-Kiebooms G, Swerts A, Cassimann JJ, et al. The motivation of at-risk individuals and their partners in deciding for or against predictive testing for Huntington’s disease. Clin Genet 1989;35:29. Sorenson SA, Fenger K. Suicide in patients with Huntington’s disease and their sibs. Am J Hum Genet 1991;49:316. Schoenfeld M, Myers RH, Cupples LA, et al. Increased rate of suicide among patients with Huntington’s disease. J Neurol Neurosurg Psychiatry 1984;47:1283. Almqvist EW, Bloch M, Brinkman R, et al., on behalf of an International Huntington Disease Collaborative Group. A worldwide assessment of the frequency of suicide, suicide attempts or psychiatric hospitalization after predictive testing for Huntington disease. Am J Hum Genet 1999;64:1293. van Duijn E, Kingma EM, van der Mast RC. Psychopathology in verified Huntington’s disease gene carriers. J Neuropsychiatry Clin Neurosci 2007;19:441. Kachian ZR, Cohen-Zimerman S, Bega D, et al. Suicidal ideation and behavior in Huntington’s disease: systematic review and recommendations. J Affect Disord 2019;250:319. McGarry A, McDermott MP, Kieburtz K, et al. Risk factors for suicidality in Huntington disease. Neurology 2019;92:e1643. Wesson M, Boileau NR, Perlmutter JS, et al. Suicidal ideation assessment in individuals with premanifest and manifest Huntington disease. J Huntingtons Dis 2018;7:239. Markel DS, Young AB, Penney JB. At-risk persons’ attitudes toward presymptomatic and prenatal testing of Huntington’s disease in Michigan. Am J Med Genet 1987;26:295. Lamport AN. Presymptomatic testing for Huntington’s chorea: ethical and legal issues. Am J Med Genet 1987;26:307. Taylor CA, Myers RH. Long-term impact of Huntington disease linkage testing. Am J Med Genet 1997:70:365.

743. Hayden MR. Predictive testing for Huntington disease: are we ready for widespread community implementation? Am J Med Genet 1991;40:515. 744. Nance MA, Leroy BS, Orr HT, et al. Protocol for genetic testing in Huntington disease: three years of experience in Minnesota. Am J Med Genet 1991;40:518. 745. Decruyenaere M, Evers-Kiebooms G, Cloostermans T, et al. Psychological distress in the 5-year period after predictive testing for Huntington’s disease. Eur J Hum Genet 2003;11:30. 746. Winnberg E, Winnberg U, Pohlkamp L, et al. What to do with a second chance in life? Long-term experiences of non-carriers of Huntington’s disease. J Genet Couns 2018;27:1438. 747. Gusella JF, McNeil S, Persichetti F, et al. Huntington’s disease. Cold Spring Harb Symp Quant Biol 1996;61:615. 748. Kremer B, Goldberg P, Andrew SE. A worldwide study of the Huntington’s disease mutation: the sensitivity and specificity of measuring CAG repeats. N Engl J Med 1994;330:1401. 749. Alonso ME, Yescas P, Rasmussen A, et al. Homozygosity in Huntington’s disease: new ethical dilemma caused by molecular diagnosis. Clin Genet 2002;61:437. 750. Evans DGR, Maher EF, Macleod R, et al. Uptake of genetic testing for cancer predisposition. J Med Genet 1997;34:746. 751. Veldhuisen B, Saris JJ, deHaij S, et al. A spectrum of mutations in the second gene for autosomal dominant polycystic kidney disease (PKD2). Am J Hum Genet 1997;61:547. 752. Deltas CC, Christodoulou K, Tjakouri C, et al. Presymptomatic molecular diagnosis of autosomal dominant polycystic kidney disease using PKD1- and PKD2-linked markers in Cypriot families. Clin Genet 1996;50:10. 753. Pirson Y, Chaveau D. Intracranial aneurysms in autosomal dominant polycystic kidney disease. In: Watson ML, Torres VE, eds. Polycystic kidney disease. Oxford: Oxford University Press, 1996:530. 754. Liu D, Wang CJ, Judge DP, et al. A Pkd1-Fbn1 genetic interaction implicates TGF-𝛽 signaling in the pathogenesis of vascular complications in autosomal dominant polycystic kidney disease. J Am Soc Nephrol 2014;25:81. 755. Sujansky E, Kreutzer SB, Johnson AM, et al. Attitudes of at-risk and affected individuals regarding presymptomatic testing for autosomal dominant polycystic kidney disease. Am J Med Genet 1990;35:510.

CHAPTER 1

Genetic Counseling: Preconception, Prenatal, and Perinatal 91

756. Hannig VL, Hopkins JR, Johnson HK, et al. Presymptomatic testing for adult onset polycystic kidney disease in at-risk kidney transplant donors. Am J Med Genet 1991;40:425. 757. Oyazato Y, Iijima K, Emi M, et al. Molecular analysis of TSC2/PKD1 contiguous gene deletion syndrome. Kobe J Med Sci 2011;57:E1. 758. Giardiello FM, Brensinger JD, Petersen GM, et al. The use and interpretation of commercial APC gene testing for familial adenomatous polyposis. N Engl J Med 1997;336:823. 759. Hogan J, Turner A, Tucker K, et al. Unintended diagnosis of Von Hippel-Lindau syndrome using Array Comparative Genomic Hybridization (CGH): counseling challenges arising from unexpected information. J Genet Couns 2013;22:22. 760. Telander RL, Zimmerman D, Sizemore GW, et al. Medullary carcinoma in children: results of early detection and surgery. Arch Surg 1989;124:841. 761. Ross LF. Predictive genetic testing for conditions that present in childhood. Kennedy Inst Ethics J 2002;12:225. 762. Wertz DC, Fanos JH, Reilly PR. Genetic testing for children and adolescents: who decides? JAMA 1994;272:875. 763. Lancaster JM, Wiserman RW, Berchuk A. An inevitable dilemma: prenatal testing for mutations in the BRCA1 breast-ovarian cancer susceptibility gene. Obstet Gynecol 1996;87:306. 764. DudokdeWit AC, Tibben A, Frets PG, et al. BRCA1 in the family: a case description of the psychological implications. Am J Med Genet 1997;71:63. 765. Julian-Reynier C, Eisinger F, Vennin P, et al. Attitudes towards cancer predictive testing and transmission of information to the family. J Med Genet 1996;33:731. 766. Lancaster JM, Wiseman RW, Berchuck A. An inevitable dilemma: prenatal testing for mutations in the BRCA1 breast-ovarian cancer susceptibility gene. Obstet Gynecol 1996;87:306. 767. Antoniou A, Pharoah PD, Narod S, et al. Average risks of breast and ovarian cancer associated with BRCA1 or BRCA2 mutations detected in case series unselected for family history: a combined analysis of 22 studies. Am J Hum Genet 2003;72:1117. 768. National Comprehensive Cancer Network. Practice guidelines in oncology. Genetic/familial high-risk assessment: breast and ovarian. Version 1. Fort Washington, PA: National Comprehensive Cancer Network, 2008. 769. Burke W, Daly M, Garber J, et al. Recommendations for follow-up care of individuals with an inherited predisposition to cancer. II. BRCA1 and

770.

771.

772.

773. 774.

775.

776.

777.

778.

779.

780.

781.

782.

783.

BRCA2. Cancer Genetics Studies Consortium. JAMA 1997;277:997. King M-C, Marks JH, Mandell JB. Breast and ovarian cancer risks due to inherited mutations in BRCA1 and BRCA2. Science 2003:302;643. Laken SJ, Petersen GM, Gruber SB, et al. Familial colorectal cancer in Ashkenazim due to a hypermutable tract in APC. Nat Genet 1997;17:79. Verkerk AJ, Pieretti M, Sutcliffe JS, et al. Identification of a gene (FMR-1) containing a CGG repeat coincident with a breakpoint cluster region exhibiting length variation in fragile X syndrome. Cell 1991;65:905. Paulson H. Repeat expansion diseases. Handb Clin Neurol 2018;147:105. Lehesjoki A-E, Kälviäinen R. Progressive myoclonic epilepsy type 1. GeneReviews [internet]. Seattle (WA): University of Washington, Seattle, 1993–2020, 2004 Jun 24 [updated 2020 Jul 2]. Ishikawa K, Dürr A, Klopstock T, et al. Pentanucleotide repeats at the spinocerebellar ataxia type 31 (SCA31) locus in Caucasians. Neurology 2011;77:1853. Corbett MA, Kroes T, Veneziano L, et al. Intronic ATTTC repeat expansions in STARD7 in familial adult myoclonic epilepsy linked to chromosome 2. Nat Commun 2019;10:4920. Campuzano V, Montermini L, Molot MD, et al. Friedreich’s ataxia: autosomal recessive disease caused by an intronic GA triplet repeat expansion. Science 1996;271:1423. Yum K, Wang ET, Kalsotra A. Myotonic dystrophy: disease repeat range, penetrance, age of onset, and relationship between repeat size and phenotypes. Curr Opin Genet Dev 2017;44:30. Jaspert A, Fahsold R, Grehl H, et al. Myotonic dystrophy: correlation of clinical symptoms with the size of the CTG trinucleotide repeat. J Neurol 1995;242:99. Schoser B. Myotonic dystrophy type 2. [internet]. Seattle (WA): UniGeneReviews versity of Washington, Seattle, 1993–2020, 2006 Sep 21 [updated 2020 Mar 19]. Higgs C, Hilbert JE, Wood L, et al. Reproductive cancer risk factors in women with myotonic dystrophy (DM): survey data from the US and UK DM registries. Front Neurol 2019;10:1071. Alsaggaf R, Pfeiffer RM, Wang Y, et al. Diabetes, metformin and cancer risk in myotonic dystrophy type 1. Int J Cancer 2020;147:785. Myers RH, MacDonald ME, Koroshetz WJ et al. De novo expansion of a (CAG)n repeat in sporadic Huntington’s disease. Ann Neurol 1992;32:707.

®

®

92 Genetic Disorders and the Fetus

784. Trottier Y, Briancalana V, Mandel JL. Instability of CAG repeats in Huntington’s disease: relation to parental transmission and age of onset. J Med Genet 1994;31:377. 785. Joosten IBT, Hellebrekers DMEI, de Greef BTA, et al. Parental repeat length instability in myotonic dystrophy type 1 pre- and protomutations. Eur J Hum Genet 2020;28(7):956. 786. Margolesky J, Starosta-Rubenstein S, Verma A, et al. A co-occurrence of trinucleotide repeat disorders. Mov Disord Clin Pract 2018;5(6):643. 787. McFarland KN, Liu J, Landrian I, et al. Paradoxical effects of repeat interruptions on spinocerebellar ataxia type 10 expansions and repeat instability. Eur J Hum Genet 2013;21:1272. 788. Cumming SA, Hamilton MJ, Robb Y, et al. De novo repeat interruptions are associated with reduced somatic instability and mild or absent clinical features in myotonic dystrophy type 1. Eur J Hum Genet 2018;26(11):1635. 789. Pešovi´c J, Peri´c S, Brkušanin M, et al. Repeat interruptions modify age at onset in myotonic dystrophy type 1 by stabilizing DMPK expansions in somatic cells. Front Genet 2018;9:601. 790. Laffita-Mesa JM, Rodriguez Pupo JM, Moreno Sera R, et al. De novo mutations in ataxin-2 gene and ALS risk. PLoS One 2013;8:e70560. 791. Beaudin M, Matilla-Dueñas A, Soon B, et al. The classification of autosomal recessive cerebellar ataxias: a consensus statement from the society for research on the cerebellum and ataxias task force. Cerebellum 2019;18(6):1098. 792. Robberecht W, Philips T. The changing scene of amyotrophic lateral sclerosis. Nat Rev Neurosci 2013;14:248. 793. Majounie E, Renton AE, Mok K, et al. Frequency of the C9orf72 hexanucleotide repeat expansion in patients with amyotrophic lateral sclerosis and frontotemporal dementia: a cross-sectional study. Lancet Neurol 2012;11:323. 794. Beck J, Poulter M, Hensman D, et al. Large C9orf72 hexanucleotide repeat expansions are seen in multiple neurodegenerative syndromes and are more frequent than expected in the UK population. Am J Hum Genet 92:345. 795. Williams KL, Fifita JA, Vucic S, et al. Pathophysiological insights into ALS with C9orf72 expansions. J Neurol Neurosurg Psychiatry 2013;84:931. 796. Lindquist SG, Duno M, Batbayli M, et al. Corticobasal and ataxia syndromes widen the spectrum of C9orf72 hexanucleotide expansion disease. Clin Genet 2013;83:279.

797. van der Zee J, Gijselinck I, Dillen L, et al. A pan-European study of the C9orf72 repeat associated with FTLD: geographic prevalence, genomic instability, and intermediate repeats. Hum Mutat 2013;34:363. 798. Devenney EM, Ahmed RM, Halliday G, et al. Psychiatric disorders in C9orf72 kindreds: study of 1,414 family members. Neurology 2018;91:e.1498. 799. Esselin F, Mouzat K, Polge A, et al. Clinical phenotype and inheritance in patients with C9ORF72 hexanucleotide repeat expansion: results from a large French cohort. Front Neurosci 2020;14:316. 800. Shi D, Xu J, Niu W, et al. Live births following preimplantation genetic testing for dynamic mutation diseases by karyomapping: a report of three cases. J Assist Reprod Genet 2020;37:539. 801. Fernández RM, Lozano-Arana MD, Sánchez B, et al. Preimplantation genetic diagnosis for myotonic dystrophy type 1 and analysis of the effect of the disease on the reproductive outcome of the affected female patients. Biomed Red Int 2017;2017:9165363. 802. Milunsky A, Baldwin C, Milunsky J. Molecular genetics and prenatal diagnosis. In: Milunsky A, Milunsky J, eds. Genetic disorders and the fetus: diagnosis, prevention and treatment, 7th edn. Hoboken, NJ: John Wiley & Sons, 2016:380. 803. Gerbrands LC, Haarman EG, Hankel MA, et al. Cystic fibrosis and Silver-Russell syndrome due to a partial maternal isodisomy of chromosome 7. Clin Case Rep 2017;5(10):1697. 804. Sotomayor FV, Abarca-Barriga HH. Homozygous deletion of the CFTR gene caused by interstitial maternal isodisomy in a Peruvian child with cystic fibrosis. J Pediatr Genet 2019;8(3):147. 805. Ammerpohl O, Martín-Subero JI, Richter J, et al. Hunting for the 5th base: techniques for analyzing DNA methylation. Biochim Biophys Acta 2009;1790:847. 806. Dedeurwaerder S, Defrance M, Calonne E, et al. Evaluation of the Infinium methylation 450K technology. Epigenomics 2011;3:771. 807. Begemann M, Rezwas FI, Beygo J, et al. Maternal variants in NLRP and other maternal effect proteins are associated with multilocus imprinting disturbance in offspring. J Med Genet 2018;55:497. 808. Yauy K, de Leeuw N, Yntema HG, et al. Accurate detection of clinically relevant uniparental disomy from exome sequencing data. Genet Med 2020;22:803. 809. Bhoj EJ, Rajabi F, Baker SW, et al. Imprinted genes in clinical exome sequencing: Review of 538 cases and exploration of mouse-human conservation in the

CHAPTER 1

810.

811.

812.

813.

814.

815.

816.

817.

818.

819.

820.

821.

822.

Genetic Counseling: Preconception, Prenatal, and Perinatal 93

identification of novel human disease loci. Eur J Med Genet 2020;63:103903. del Gaudio D, Shinawi M, Astbury C, et al. Diagnostic testing for uniparental disomy: a points to consider statement from the American College of Medical Genetics and Genomics (ACMG). Genet Med 2020;22:1133. Court F, Martin-Trujillo A, Romanelli V, et al. Genome-wide allelic methylation analysis reveals disease-specific susceptibility to multiple methylation defects in imprinting syndromes. Hum Mutat 2013;34:595. Elbracht M, Mackay D, Begemann M, et al. Disturbed genomic imprinting and its relevance for human reproduction: causes and clinical consequences. Hum Reprod Update 2020;26:197. Docherty LE, Rezwan FI, Poole RL, et al. Mutations in NLRP5 are associated with reproductive wastage and multilocus imprinting disorders in humans. Nat Commun 2015;6:8086. Soellner L, Begemann M, Degenhardt F, Geipel A, et al. Maternal heterozygous NLRP7 variant results in recurrent reproductive failure and imprinting disturbances in the offspring. Eur J Hum Genet 2017;25:924. Lee MT, Bonneau AR, Giraldez AJ. Zygotic genome activation during the maternal-to-zygotic transition. Annu Rev Cell Dev Biol 2014;30:581. Zhou LQ, Dean J. Reprogramming the genome to totipotency in mouse embryos. Trends Cell Biol 2015;25:82. Vicenz C, Lovett JL, Wu W, et al. Loss of imprinting in human placentas is widespread, coordinated, and predicts birth phenotypes. Mol Biol Evol 2020;37:429. Giabicani É, Brioude F, Le Bouc Y, et al. Imprinted disorders and growth. Ann Endocrinol (Paris) 2017;78:112. Rhon-Calderon EA, Vrooman LA, Riesche L, et al. The effects of assisted reproductive technologies on genomic imprinting in the placenta. Placenta 2019;84:37. Litzky JP, Deyssenroth MA, Everson TM, et al. Prenatal exposure to maternal depression and anxiety on imprinted gene expression in placenta and infant neurodevelopment and growth. Pediatr Res 2018;83(5):1075. John RM. Imprinted genes and the regulation of placental endocrine function: pregnancy and beyond. Placenta 2017;56:86. Hattori H, Hiura H, Kitamura A, et al. Association of four imprinting disorders and ART. Clin Epigenetics 2019;11:21.

823. Uk A, Collardeau-Frachon S, Scanvion Q, et al. Assisted reproductive technologies and imprinting disorders: results of a study from a French congenital malformations registry. Eur J Med Genet 2018;61:518. 824. Mackay DJG, Temple IK. Human imprinting disorders: principles, practice, problems, and progress. Eur J Med Genet 2017;60:618. 825. Henningsen AA, Gissler M, Rasmussen S, et al. Imprinting disorders in children born after ART: a Nordic study from CoNARTaS group. Hum Reprod 2020;35:1178. 826. Johnson JP, Beischel L, Schwanke C, et al. Overrepresentation of pregnancies conceived by artificial reproductive technology in prenatally identified fetuses with Beckwith-Wiedemann syndrome. J Assist Reprod Genet 2018;35:985. 827. Mussa A, Molinatto C, Cerrato F, et al. Assisted reproductive techniques and risk of Beckwith-Wiedemann syndrome. Pediatrics 2017;140:e20164311. 828. Cortessis VK, Azadian M, Buxbaum J, et al. Comprehensive meta-analysis reveals association between multiple imprinting disorders and conception by assisted reproductive technology. J Assist Reprod Genet 2018;35:943. 829. Monk D, Mackay DJG, Eggermann T, et al. Genomic imprinting disorders: lessons on how genome, epigenome and environment interact, Nat Rev Genet 2019;20:235. 830. Court F, Tayama C, Romanelli V, et al. Genome-wide parent-of-origin DNA methylation analysis reveals the intricacies of human imprinting and suggests a germline methylation-independent mechanism of establishment. Genome Res 2014;24:554. 831. Weksberg R, Shuman C, Caluseriu O, et al. Discordant KCNQ1OT1 imprinting in sets of monozygotic twins discordant for Beckwith-Wiedemann syndrome. Hum Mol Genet 2002;11:1317. 832. Bliek J, Alders M, Maas SM, et al. Lessons from BWS twins: complex maternal and paternal hypomethylation and a common source of haematopoietic stem cells. Eur J Hum Genet 2009;17:1625. 833. Riess A, Binder G, Ziegler J, et al. First report on concordant monozygotic twins with Silver-Russell syndrome and ICR1 hypomethylation. Eur J Med Genet 2016;59:1. 834. DeAngelis AM, Martini AE, Owen CM. Assisted reproductive technology and epigenetics. Semin Reprod Med 2018;36:221. 835. Bragin E, Chatzimichali EA, Wright CF, et al. DECIPHER: database for the interpretation of phenotype-linked plausibly pathogenic sequence

94 Genetic Disorders and the Fetus

836.

837.

838.

839.

840.

841.

842.

843.

844.

845.

846.

847.

848.

849.

and copynumber variation. Nucleic Acids Res 2014;42(Database issue):D993. Johnston JJ, Biesecker LG. Databases of genomic variation and phenotypes: existing resources and future needs. Hum Mol Genet 2013;22(R1):R27. Lu J, Campeau P, Lee B. Genotype-phenotype correlation – promiscuity in the era of next generation sequencing. N Engl J Med 2014;371:593. Adam MP, Banka S, Bjornsson HT, et al. Kabuki syndrome: international consensus diagnostic criteria. J Med Genet 2019;56:89. Bögershausen N, Gatinois V, Riehmer V, et al. Mutation update for Kabuki syndrome genes KMT2D and KDM6A and further delineation of X-linked Kabuki syndrome subtype 2. Hum Mutat 2016;37:847. Shangguan H, Su C, Ouyang Q, et al. Kabuki syndrome: novel pathogenic variants, new phenotypes and review of the literature. Orphanet J Rare Dis 2019;14:255. Banka S, Lederer D, Benoit V, et al. Novel KDM6A (UTX) mutations and a clinical and molecular review of the X-linked Kabuki syndrome (KS2). Clin Genet 2015;87:252. Yap KL, Johnson AEK, Fischer D, et al. Correction: “Congenital hyperinsulinism as the presenting feature of Kabuki syndrome: clinical and molecular characterization of 10 affected individuals”. Genet Med 2019;21:262. Courcet JB, Faivre L, Michot C, et al. Clinical and molecular spectrum of renal malformations in Kabuki syndrome. J Pediatr 2013;163:742. Pyeritz RE, McKusick VA. The Marfan syndrome: diagnosis and management. N Engl J Med 1979;300:772. Beutler E, Nguyen NJ, Henneberger MW, et al. Gaucher disease: gene frequencies in the Ashkenazi Jewish population. Am J Hum Genet 1993;53:85. Lewis BD, Nelson PV, Robertson EF, et al. Mutation analysis of 28 Gaucher disease patients: the Australasian experience. Am J Med Genet 1994;49:218. Kerem E, Corey M, Kerem B, et al. The relationship between genotype and phenotype in cystic fibrosis: analysis of the most common mutation. N Engl J Med 1990;323:1517. Cystic Fibrosis Genotype-Phenotype Consortium. Correlation between genotype and phenotype in patients with cystic fibrosis. N Engl J Med 1993;329:1308. Dork T, Wulbrand U, Richter T, et al. Cystic fibrosis with three mutations in the cystic fibrosis transmembrane regulator gene. Hum Genet 1991;87:441.

850. Le C, Ramjeesingh M, Reys E, et al. The cystic fibrosis mutation (F508) does not influence the chloride channel activity of CFTR. Nat Genet 1993;3:311. 851. Cutting GR. Modifier genes in Mendelian disorders: the example of cystic fibrosis. Ann NY Acad Sci 2010;1214:57. 852. Bradley GM, Blackman SM, Watson CP, et al. Genetic modifiers of nutritional status in cystic fibrosis. Am J Clin Nutr 2012;96:1299. 853. Blackman SM, Commander CW, Watson C. Genetic modifiers of cystic fibrosis-related diabetes. Diabetes 2013;62:3627. 854. Tuddenham EGD. Factor VIII and haemophilia A. Baillière’s Clin Haematol 1989;2:849. 855. McVey JH, Rallapalli PM, Kemball-Cook G, et al. The European association for haemophilia and allied disorders (EAHAD) coagulation factor variant databases: important resources for haemostasis clinicians and researchers. Haemophilia 2020;26:306. 856. Atik T, I¸sik E, Onay H, et al. Factor 8 gene mutation spectrum of 270 patients with haemophilia A: identification of 36 novel mutations. Turk J Haematol 2020;37(3):145. 857. Miller DS, Steinbrecher RA, Wieland K, et al. The molecular genetic analysis of haemophilia A: characterization of six partial deletions in the factor VIII gene. Hum Genet 1990;86:219. 858. Sarri CA, Roussaki-Schulze A, Vasilopoulos Y, et al. Netherton syndrome: a genotype-phenotype review. Mol Diagn Ther 2017;21:137. 859. Koczkowska M, Callens T, Chen Y, et al. Clinical spectrum of individuals with pathogenic NF1 missense variants affecting p. Met1149, p.Arg1276, and p.Lys1423: genotype-phenotype study in neurofibromatosis type 1. Hum Mutat 2020;41:299. 860. Lehtonen HJ, Sipponen T, Tojander S, et al. Segregation of a missense variant in entire smooth muscle actin gamma-2 with autosomal dominant familial visceral myopathy. Gastroenterology 2012;143:1482. 861. Berdon W, Baker D, Blanc W, et al. Megacystismicrocolon-intestinal hypoperistalsis syndrome: a new cause of intestinal obstruction in the newborn. Report of radiologic findings in five newborn girls. AJR Am J Roentgenol 1976;126:957. 862. Puri P, Shinkai M. Megacystis-microcolon-intestinalhypoperistalsis syndrome. Semin Pediatr Surg 2005;14:58. 863. Milunsky M, Baldwin C, Zhang X, et al. Diagnosis of chronic intestinal pseudo-obstruction & megacystis by sequencing the ACTG2 gene. J Pediatr Gastroenterol Nutr 2017;65:384.

CHAPTER 1

Genetic Counseling: Preconception, Prenatal, and Perinatal 95

864. Milunsky A, Lazier J, Baldwin C, et al. Prenatal

878. Donnai D, Read AP, McKeown C, et al. Hypome-

diagnosis of chronic intestinal pseudo-obstruction

lanosis of Ito-A manifestation of mosaicism or

and paternal somatic mosaicism for the ACTG2

chimerism. J Med Genet 1988;25:809. 879. Thomas IT, Frias JL, Cantu ES, et al. Association

pathogenic variant. Prenat Diagn 2017;37:1254. 865. Xiromerisiou G, Marogianni C, Dadouli K, et al.

of pigmentary anomalies with chromosomal and

Cerebral autosomal dominant arteriopathy with sub-

genetic mosaicism and chimerism. Am J Hum Genet

cortical infarcts and leukoencephalopathy revisited: genotype-phenotype correlations of all published

1989;45:193. 880. Raghunath M, Mackay K, Dalgleish R, et al. Genetic counseling on brittle grounds: recurring osteogenesis

cases. Neurol Genet 2020;6:e434. 866. Zhang L, Dong X, Lee M, et al. Single-cell whole-genome sequencing reveals the functional

imperfecta due to parental mosaicism for a dominant mutation. Eur J Pediatr 1995;154:123.

landscape of somatic mutations in B lymphocytes

881. Lund AM, Nicholls AC, Schwartz M, et al. Parental

across human lifespan. Proc Natl Acad Sci U S A

mosaicism and autosomal dominant mutations caus-

2019;116:9014.

ing structural abnormalities of collagen I are frequent

867. Vijg J. Somatic mutations, genome mosaicism, cancer

in families with osteogenesis imperfecta type III/IV. Acta Paediatr 1997;86:711.

and aging. Curr Opin Genet Dev 2014;26:141. al.

882. Telenius H, Kremer B, Goldberg YP, et al. Somatic

PIK3CA-associated developmental disorders exhibit

and gonadal mosaicism of the Huntington disease

distinct classes of mutations with variable expression

gene CAG repeat in brain and sperm. Nat Genet

868. Mirzaa

G,

Timms

AE,

Conti

V,

et

and tissue distribution. JCI Insight 2016;1:e.87623.

1994;6:409.

869. Lalonde E, Ebrahimzadeh J, Rafferty K, et al. Molec-

883. Cancel G, Durr A, Didierjean O, et al. Molecular

ular diagnosis of somatic overgrowth conditions: a

and clinical correlations in spinocerebellar ataxia 2:

singular-center experience. Mol Genet Genomic Med 2019;7:e536.

a study of 32 families. Hum Mol Genet 1997;6:709. 884. Satge D, Geneix A, Goburdhun J, et al. A history of

870. Quinlan-Jones E, Williams D, Bell C, et al. Prena-

miscarriages and mild prognathism as possible mode

tal detection of PIK3CA-related overgrowth spec-

of presentation of mosaic trisomy 18 in women. Clin

trum in cultured amniocytes using long-range PCR and next-generation sequencing. Pediatr Dev Pathol 2017;20(1):54. 871. Tung JY, Lai SHY, Au SLK, et al. Coexistence of paternally-inherited ABCC8 mutation and mosaic paternal uniparental disomy 11p hyperinsulinism. Int J Pediatr Endocrinol 2020;2020:13. 872. Koh HY, Lee JH. Brain somatic mutations in epileptic disorders. Mol Cells 2018;41:881. 873. Liu AJ, Yang XX, Wu QX, et al. Study on mosaicism

Genet 1996;50:470. 885. Comstock JM, Putnam AR, Sangle N, et al. Recurrence of achondrogenesis type 2 in sibs: additional evidence for germline mosaicism. Am J Med Genet A 2010;152A;1822. 886. Abe J, Nakamura K, Nishikomori R, et al. A nationwide survey of Aicardi-Goutières syndrome patients identifies a strong association between dominant TREX1 mutations and chilblain lesions: Japanese cohort study. Rheumatology (Oxford) 2014;53:448.

of SCN1A gene mutation in parents of children

887. Bruttini M, Vitelli F, Meloni I, et al. Mosaicism in

with Dravet syndrome. Zhonghua Er Ke Za Zhi

Alport syndrome with genetic counselling. J Med

2017;55:818.

Genet 2000;37:717.

874. Dehainault C, Golmard L, Millot GA, et al. Mosaicism

888. Erickson RP. Somatic gene mutation and human

and prenatal diagnosis options: insights from

disease other than cancer: an update. Mutat Res

retinoblastoma. Eur J Hum Genet 2017;25:381.

2010;705:96.

875. D’Gama AM, Walsh CA. Somatic mosaicism and neu-

889. Meira JGC, Sarno MAC, Faria ÁCO, et al. Diagno-

rodevelopmental disease. Nat Neurosci 2018;21:1504.

sis of Atelosteogenesis type I suggested by fetal ultra-

876. Rodin RE, Walsh CA. Somatic mutation in pediatric

sonography and atypical paternal phenotype with

neurological diseases. Pediatr Neurol 2018;87:20.

mosaicism. Rev Bras Ginecol Obstet 2018;40(9):570.

877. Pham J, Shaw C, Pursley A, et al. Somatic mosaicism

890. Haack TB, Hogarth P, Kruer MC, et al. Exome

detected by exon-targeted, high resolution aCGH

sequencing reveals de novo WDR45 mutations caus-

in 10362 consecutive cases. Eur J Hum Genet

ing a phenotypically distinct, X-linked dominant

2014;22:969.

form of NBIA. Am J Hum Genet 2012;91:1144.

96 Genetic Disorders and the Fetus

891. Roux-Buisson N, Egéa G, Denjoy I, et al. Germline and somatic mosaicism for a mutation of the ryanodine receptor type 2 gene: implication for genetic counseling and patient caring. Europace 2011;13:120. 892. Catteruccia M, Fattori F, Codemo V, et al. Centronuclear myopathy related to dynamin 2 mutations: clinical, morphological, muscle imaging and genetic features of an Italian cohort. Neuromuscul Disord 2013;23:229. 893. Taioli F, Bertolasi L, Ajena D, et al. Parental mosaicism of a novel PMP22 mutation with minimal neuropathic phenotype. J Peripher Nerv Syst 2012;17:414. 894. Omoyinmi E, Melo Gomes S, Standing A, et al. Brief report: whole-exome sequencing revealing somatic NLRP3 mosaicism in a patient with chronic infantile neurologic, cutaneous, articular syndrome. Arthritis Rheumatol 2014;66:197. 895. Tanaka N, Izawa K, Saito MK, et al. High incidence of NLRP3 somatic mosaicism in patients with chronic infantile neurologic, cutaneous, articular syndrome: results of an International Multicenter Collaborative Study. Arthritis Rheum 2011;63:3625. 896. Nagendran S, Richards AJ, McNinch A, et al. Somatic mosaicism and the phenotype expression of COL2A1 mutations. Am J Med Genet A 2012;158A:1204. 897. Meguro T, Yoshida Y, Hayashi M, et al. Inheritance of polyalanine expansion mutation of PHOX2B in congenital central hypoventilation syndrome. J Hum Genet 2012;57:335. 898. Ng BG, Buckingham KJ, Raymond K, et al. Mosaicism of the UDP-galactose transporter SLC35A2 causes a congenital disorder of glycosylation. Am J Hum Genet 2013;92:632. 899. Kurek KC, Luks VL, Ayturk UM, et al. Somatic mosaic activating mutations in PIK3CA cause CLOVES syndrome. Am J Hum Genet 2012;90:1108. 900. Baquero-Montoya C, Gil-Rodríguez MC, Braunholz D, et al. Somatic mosaicism in a Cornelia de Lange syndrome patient with NIPBL mutation identified by different next generation sequencing approaches. Clin Genet 2014;86:595. 901. Girisha KM, Lewis LE, Phadke SR, et al. Costello syndrome with severe cutis laxa and mosaic HRAS G12S mutation. Am J Med Genet A 2010;152A:2861. 902. Alzualde A, Moreno F, Martinez-Lage P, et al. Somatic mosaicism in a case of apparently sporadic Creutzfeldt-Jakob disease carrying a de novo D178n mutation in the PRNP gene. Am J Med Genet B Neuropsychiatr Genet 2010;153B:1283. 903. Goriely A, Lord H, Lim J, et al. Germline and somatic mosaicism for FGFR2 mutation in the mother of

904.

905.

906.

907.

908.

909.

910.

911.

912.

913.

914.

915.

916.

a child with Crouzon syndrome: implications for genetic testing in “paternal age-effect” syndromes. Am J Med Genet A 2010;152:2067. Zampatti S, Mela J, Peconi C, et al. Identification of Duchene/Becker muscular dystrophy mosaic carriers through a combined DNA/RNA analysis. Prenat Diagn 2018;38(13):1096. Filho AB, Souza J, Faucz FR, et al. Somatic/gonadal mosaicism in a syndromic form of ectrodactyly, including eye abnormalities, documented through array-based comparative genomic hybridization. Am J Med Genet A 2011;155A:1152. Bourdeaut F, Hérault A, Gentien D, et al. Mosaicism for oncogenic G12D KRAS mutation associated with epidermal nevus, polycystic kidneys and rhabomyosarcoma. J Med Genet 2010;47:859. Terracciano A, Specchio N, Darra F, et al. Somatic mosaicism of PCDH19 mutation in a family with low-penetrance EFMR. Neurogenetics 2012;13:341. Dibbens LM, Kneen R, Bayly MA, et al. Recurrence risk of epilepsy and mental retardation in females due to parental mosaicism of PCDH19 mutations. Neurology 2011;76:1514. Maclellan RA, Luks VL, Vivero MP, et al. PIK3CA activating mutations in facial infiltrating lipomatosis. Plast Reconstr Surg 2014;133:12e. Jansen AC, Oostra A, Desprechins B, et al. TUBA1A mutations: from isolated lissencephaly to familial polymicrogyria. Neurology 2011;76:988. Pinto FO, Leblanc T, Chamousset D, et al. Diagnosis of Fanconi anemia in patients with bone marrow failure. Haematologica 2009;94:487. Li X, Jiang M, Han W, et al. A novel TNNI2 mutation causes Freeman-Sheldon syndrome in a Chinese family with an affected adult with only facial contractures. Gene 2013;527:630. Filipe B, Albuquerque Q, Bik E, et al. APC somatic mosaicism in a patient with Gardner syndrome carrying the E1573X mutation: report of a case. Dis Colon Rectum 2009;52:1516. Lee JH, Huynh M, Silhavy JL, et al. De novo somatic mutations in components of the PI3K-AKT3-mTOR pathway cause of hemi-megalencephaly. Nat Genet 2012;44:941. Eyries M, Coulet F, Girerd B, et al. ACVRL1 germinal mosaic with two mutant alleles in hereditary hemorrhagic telangiectasia associated with pulmonary arterial hypertension. Clin Genet 2012;82:173. Pastrello C, Fornasarig M, Pin E, et al. Somatic mosaicism in a patient with Lynch syndrome. Am J Med Genet A 2009;149A:212.

CHAPTER 1

Genetic Counseling: Preconception, Prenatal, and Perinatal 97

917. Hsu AP, Sowerwine KJ, Lawrence MG, et al. Intermediate phenotypes in patients with autosomal dominant hyper-IgE syndrome caused by somatic mosaicism. J Allergy Clin Immunol 2013;131:1586. 918. Rabin R, Millan F, Cabrera-Luque J, et al. Intellectual disability due to monoallelic variant in GATAD2B and mosaicism in unaffected parent. Am J Med Genet A 2018;176(12):2907. 919. Tsubahara M, Hayashi Y, Niijima S, et al. Isolated growth hormone deficiency in two siblings because of paternal mosaicism for a mutation in the GH1 gene. Clin Endocrinol (Oxf) 2012;76:420. 920. Doisaki S, Muramatsu H, Shimada A, et al. Somatic mosaicism for oncogenic NRAS mutations in juvenile myelomonocytic leukemia. Blood 2012;120:1485. 921. Hafner C, Toll A, Gantner S, et al. Keratinocyte epidermal nevi are associated with mosaic RAS mutations. J Med Genet 2012;49:249. 922. Prochazkova K, Pavlikova K, Minarik M, et al. Somatic TP53 mosaicism in a patient with Li-Fraumeni syndrome. Am J Med Genet A 2009;149A:206. 923. Thibodeau IL, Xu J, Li Q, et al. Paradigm of genetic mosaicism and lone atrial fibrillation: physiological characterization of a connexin 43-deletion mutant identified from atrial tissue. Circulation 2010;122:236. 924. Amary MF, Damato S, Halai D, et al. Ollier disease and Maffucci syndrome are caused by somatic mosaic mutations of IDH1 and IDH2. Nat Genet 2011;43:1262. 925. Vissers LE, Fano V, Martinelli D, et al. Whole-exome sequencing detects somatic mutations of IDH1 in metaphyseal chondromatosis with D-2-hydroxyglutaric aciduria (MC-HGA). Am J Med Genet A 2011;155A:2609. 926. Odgerel Z, Sarkozy A, Lee HS, et al. Inheritance patterns and phenotypic features of myofibrillar myopathy associated with a BAG3 mutation. Neuromuscul Disord 2010;20:438. 927. Catalli C, Morgante A, Iraci R, et al. Validation of sensitivity and specificity of tetraplet-primed PCR (TP-PCR) in the molecular diagnosis of myotonic dystrophy type 2 (DM2). J Mol Diagn 2010;12:601. 928. Marini M, Bocciardi R, Gimelli S, et al. A spectrum of LMX1B mutations in Nail-Patella syndrome: new point mutations, deletion, and evidence of mosaicism in unaffected parents. Genet Med 2010;12:431. 929. Messiaen L, Vogt J, Bengesser K, et al. Mosaic type-1 NF1 microdeletions as a cause of both generalized and segmental neurofibromatosis type-1 (NF1). Hum Mutat 2011;32:213.

930. Goutagny S, Bah AB, Parfait B, et al. Neurofibromatosis type 2 in the elderly population: clinical and molecular features. Am J Med Genet A 2013;161A:667. 931. Saitsu H, Hoshino H, Kato M, et al. Paternal mosaicism of an STXBP1 mutation in OS. Clin Genet 2011;80:484. 932. Szuhai K, Jennes I, de Jong D, et al. Tiling resolution array-CGH shows that somatic mosaic deletion of the EXT gene is causative in EXT gene mutation negative multiple osteochondromas patients. Hum Mutat 2011;32:e2063. 933. Hague J, Delon I, Brugger K, et al. Male child with somatic mosaic Osteopathia Striata with Cranial Sclerosis caused by a novel pathogenic AMER1 frameshift mutation. Am J Med Genet A 2017;173(7):1931. 934. Kalish JM, Conlin LK, Mostoufi-Moab S, et al. Bilateral pheochromocytomas, hemihyperplasia, and subtle somatic mosaicism: the importance of detecting low-level uniparental disomy. Am J Med Genet A 2013;161A:993. 935. Steinbusch CV, van Roozendaal KE, Tserpelis D, et al. Somatic mosaicism in a mother of two children with Pitt-Hopkins syndrome. Clin Genet 2013;83:73. 936. Buffet A, Smati S, Mansuy L, et al. Mosaicism in HIF2A-related polycythemia-paraganglioma syndrome. J Clin Endocrinol Metab 2014;99:e369. 937. Doubaj Y, De Sandre-Giovannoli A, Vera EV, et al. An inherited LMNA gene mutation in atypical Progeria syndrome. Am J Med Genet A 2012;158A:2881. 938. Lindhurst MJ, Sapp JC, Teer JK, et al. A mosaic activating mutation in AKT1 associated with the Proteus syndrome. N Engl J Med 2011;365:611. 939. Ngai YF, Chijiwa C, Mercimek-Mahmutoglu S, et al. Pseudohypoparathyroidism type 1a and the GNAS p.R231H mutation: somatic mosaicism in a mother with two affected sons. Am J Med Genet A 2010;152A:2784. 940. Coughlin CR 2nd, Krantz ID, Schmitt ES, et al. Somatic mosaicism for PDHA1 mutation in a male with pyruvate dehydrogenase complex deficiency. Mol Genet Metab 2010;100:296. 941. Jin ZB, Gu F, Matsuda H, et al. Somatic and gonadal mosaicism in X-linked retinitis pigmentosa. Am J Med Genet A 2007;143A:2544. 942. Rushlow D, Piovesan B, Zhang K, et al. Detection of mosaic RB1 mutations in families with retinoblastoma. Hum Mutat 2009;30:842. 943. Psoni S, Sofocleous C, Traeger-Synodinos, et al. Phenotypic and genotypic variability in four males with MECP2 gene sequence aberrations including a novel deletion. Pediatr Res 2010;67:551.

98 Genetic Disorders and the Fetus

944. Bartnik M, Derwi´nska K, Gos M, et al. Early-onset seizures due to mosaic exonic deletions of CDKL5 in a male and two females. Genet Med 2011;13:447. 945. Bartsch O, Kress W, Kempf O, et al. Inheritance and variable expression in Rubinstein-Taybi syndrome. Am J Med Genet A 2010;152A:2254. 946. Chiang PW, Lee NC, Chien N. Somatic and germ-line mosaicism in Rubinstein-Taybi syndrome. Am J Med Genet A 2009;149A:1463. 947. Carmignac V, Thevenon J, Adès L, et al. In-frame mutations in exon 1 of SKI cause of dominant Shprintzen-Goldberg syndrome. Am J Hum Genet 2012;91:950. 948. Castronovo C, Rusconi D, Crippa M, et al. A novel mosaic NSD1 intragenic deletion in a patient with an atypical phenotype. Am J Med Genet A 2013;161A:611. 949. Désir J, Cassart M, Donner C, et al. Spondyloperipheral dysplasia as the mosaic form of platyspondylic lethal skeletal dysplasia Torrance type in mother and fetus with the same COL2A1 mutation. Am J Med Genet A 2012;158A:1948. 950. Mineyko A, Doja A, Hurteau J, et al. A novel missense mutation in LIS1 in a child with subcortical band heterotopia and pachygyria inherited from his mildly affected mother with somatic mosaicism. J Child Neurol 2010;25:738. 951. Isodor B, Capito C, Paris F, et al. Familial frameshift SRY mutation inherited from a mosaic father with testicular dysgenesis syndrome. J Clin Endocrinol Metab 2009;94:3467. 952. Dufendach KA, Giudicessi JR, Boczek NJ, et al. Maternal mosaicism confounds the neonatal diagnosis of type 1 Timothy syndrome. Pediatrics 2013;131:e1991. 953. Inbar-Feigenberg M, Choufani S, Cytrynbaum C, et al. Mosaicism for genome-wide paternal uniparental disomy with features of multiple imprinting disorders: diagnostic and management issues. Am J Med Genet A 2013;161A:13. 954. Davis BR, Candotti F. Revertant somatic mosaicism in the Wiskott-Aldrich syndrome. Immunol Res 2009;44:127. 955. Kawai T, Nishikomori R, Izawa K, et al. Frequent somatic mosaicism of NEMO in T cells of patients with X-linked anhidrotic ectodermal dysplasia with immunodeficiency. Blood 2012;119:5458. 956. Yamada M, Okura Y, Suzuki, et al. Somatic mosaicism in two unrelated patients with X-linked chronic granulomatous disease characterized by the presence of a small population of normal cells. Gene 2012;497:110.

957. Twigg SR, Matsumoto K, Kidd AM, et al. The origin of EFNB1 mutations in craniofrontonasal syndrome: frequent somatic mosaicism and explanation of the paucity of carrier males. Am J Hum Genet 2006;78:999. 958. Cheillan D, Joncquel-Chevalier Curt M, Briand G, et al. Screening for primary creatine deficiencies in French patients with unexplained neurological symptoms. Orphanet J Rare Dis 2012;7:96. 959. Chen XL, Zhao Y, Ke HP, et al. Detection of somatic and germline mosaicism for the LAMP2 gene mutation c.808dupG in a Chinese family with Danon disease. Gene 2012;507:174. 960. Juan-Mateu J, Paradas C, Olivé M, et al. Isolated cardiomyopathy caused by a DMD nonsense mutation in somatic mosaicism: genetic normalization in skeletal muscle. Clin Genet 2012;82:574. 961. Vreeburg M, van Geel M, van den Heuij LG, et al. Focal dermal hypoplasia in a male patient due to mosaicism for a novel PORCN single nucleotide deletion. J Eur Acad Dermatol Venereol 2011;25:592. 962. Maas SM, Lombardi MP, van Essen AJ, et al. Phenotype and genotype in 17 patients with Goltz-Gorlin syndrome. J Med Genet 2009;46:716. 963. Margari L, Lamanna AL, Buttiglione M, et al. Long-term follow-up of neurological manifestations in a boy with incontinentia pigmenti. Eur J Pediatr 2013;172:1259. 964. Donsante A, Johnson P, Jansen LA, et al. Somatic mosaicism in Menkes disease suggests choroid plexus-mediated copper transport to the developing brain. Am J Med Genet A 2010;152A:2529. 965. Chénier S, Noor A, Dupuis L, et al. Osteopathia striata with cranial sclerosis and developmental delay in a male with mosaic deletion in chromosome region Xq11.2. Am J Med Genet A 2012;158A:2946. 966. Oegema R, Hulst JM, Theuns-Walks SD, et al. Novel no-stop FLNA mutation causes multi-organ involvement in males. Am J Med Genet A 2013;161:2376. 967. Ducamp S, Schneider-yin X, de Rooij F, et al. Molecular and functional analysis of the C-terminal region of human erythroid-specific 5-aminolevulinic synthase associated with X-linked dominant protoporphyria (XLDPP). Hum Mol Genet 2013;22:1280. 968. Quélin C, Saillour Y, Souville I, et al. Mosaic DCX deletion causes subcortical band heterotopia in males. Neurogenetics 2012;13:367. 969. Ptacek JT, Eberhardt TL. Breaking bad news. JAMA 1996;276:496. 970. Luz R, George A, Spitz E, et al. Breaking bad news in prenatal medicine: a literature review. J Reprod Infant Psychol 2017;35:14-31.

CHAPTER 1

Genetic Counseling: Preconception, Prenatal, and Perinatal 99

971. Bond CF, Anderson EL. The reluctance to transmit bad news: private discomfort or public display? J Eur Soc Psychol 1987;23:176. 972. Monaghan KG, Leach NT, Pekarek D, et al. The use of fetal exome sequencing in the prenatal diagnosis: a points to consider document of the American College of Medical Genetics and Genomics (ACMG). Genet Med 2020;22:675. 973. Horn R, Parker M. Opening Pandora’s box?: ethical issues in prenatal whole genome and exome sequencing. Prenat Diagn 2018;38:20. 974. Parker M, Lucassen A. Using a genetic test result in the care of family members: How does the duty of confidentiality apply? Eur J Hum Genet 2018;26:955. 975. Robyr R, Bernard JP, Roume J, et al. Familial diseases revealed by a fetal anomaly. Prenat Diagn 2006;26:1224. 976. Donnelly JC, Platt LD, Rebarber, A, et al. Association of copy number variants with specific ultrasonographically detected fetal anomalies. Obstet Gynecol 2014;124:83. 977. Wapner RJ, Martin CL, Levy B, et al. Chromosomal microarray versus karyotyping for prenatal diagnosis. N Engl J Med 2012;367:2175. 978. Hawkins A, Stenzel A, Taylor J, et al. Variables influencing pregnancy termination following prenatal diagnosis of fetal chromosome abnormalities. J Genet Couns 2013;22:238. 979. Blakeley C, Smith DM, Johnstone ED, et al. Parental decision-making following a prenatal diagnosis that is lethal, life-limiting, or has long term implications for the future child and family: a meta-synthesis of qualitative literature. BMC Med Ethics 2019;20:56. 980. Supiano K, Vaughn-Cole B. The impact of personal loss on the experience of health professions: graduate students in end-of life and bereavement care. Death Stud 2011;35:73. 981. Silver J, Caleshu C, Casson-Parkin S, et al. Mindfulness among genetic counselors is associated with increased empathy and work engagement and decreased burnout and compassion fatigue. J Genet Couns 2018;27:1175. 982. Verp M, Bombard A, Simpson J, et al. Parental decision following prenatal diagnosis of fetal chromosome abnormality. Am J Med Genet 1988;29:613. 983. Meryash D, Abuelo D. Counselling needs and attitudes toward prenatal diagnosis and abortion in fragile X families. Clin Genet 1988;33:349. 984. Sandelowski M, Barroso J. The travesty of choosing after positive prenatal diagnosis. J Obstet Gynecol Neonatal Nurs 2005;34(3):307.

985. Lou S, Jensen LG, Peterson OB, et al. Parental response to severe or lethal prenatal diagnosis: a systematic review of qualitative studies. Prenat Diagn 2017;37(8):731. 986. Chow EWC, Watson M, Young DA, et al. Neurocognitive profile in 22q11 deletion syndrome and schizophrenia. Schizophr Res 2006;87:270. 987. Evers LJ, De Die-Smulders CE, Smeets EE, et al. The velo-cardio-facial syndrome: the spectrum of psychiatric problems and cognitive deterioration at adult age. Genet Couns 2009;20:307. 988. Baker K, Vorstman JAS. Is there a core neuropsychiatric phenotype in 22q11.2 deletion syndrome? Curr Opin Neurol 2012;25:131. 989. Murphy KC. Annotation: velo-cardio-facial syndrome. J Child Psychol Psychiat 2005;46:563. 990. Murphy KC. Schizophrenia and velo-cardio-facial syndrome. The Lancet 2002;359:426. 991. McDonald-McGinn DM, Hain HS, et al. 22q11.2 Deletion syndrome. In: Adam MP, Ardinger HH, Pagon RA, et al., eds. GeneReviews [internet]. Seattle (WA): University of Washington, Seattle, 1993–2020, September 23, 1999. 992. White-van Mourik MCA, Connor JM, Ferguson-Smith MA. The psychosocial sequelae of a second-trimester termination of pregnancy for fetal abnormality. Prenat Diagn 1992;12:189. 993. Blumberg BD, Golbus MC, Hanson K. The psychological sequelae of abortion performed for a genetic indication. Am J Obstet Gynecol 1975;122:799. 994. Blumberg BD. The emotional implications of prenatal diagnosis. In: Emery, AEH, Pullen IM, eds. Psychological aspects of genetic counselling. London: Academic Press, 1984:202. 995. October T, Dryden-Palmer K, Copnell B, et al. Caring for parents after the death of a child. Pediatr Crit Care Med 2018;19(8S Suppl 2):S61. 996. Udipi S, Veach PM, Kao J, et al. The psychic costs of empathic engagement: personal and demographic predictors of genetic counselor compassion fatigue. J Genet Couns 2008:17;459. 997. Parkes CM. Bereavement. Studies of grief in adult life. London: Tavistock Publications, 1972. 998. Worden JW. Grief counseling and grief therapy, 2nd edn. New York: Springer, 1991. 999. Cacciatore J. Psychological effects of stillbirth. Semin Fetal Neonatal Med 2013;18:76. 1000. Appleton R, Gibson B, Hey E. The loss of a baby at birth: the role of the bereavement officer. Br J Obstet Gynaecol 1993;100:51. 1001. Seller M, Barnes C, Ross S, et al. Grief and midtrimester fetal loss. Prenat Diagn 1993;13:341.

®

100 Genetic Disorders and the Fetus

1002. Fanos JH. Developmental tasks of childhood and adolescence: implications for genetic testing. Am J Med Genet 1997;71:22. 1003. Wertz DC, Fanos JH, Reilly PR. Genetic testing for children and adolescents: who decides? JAMA 1994;272:875. 1004. Clinical Genetics Society (UK). Report of a working party: the genetic testing of children. J Med Genet 1994;31:785. 1005. American Society of Human Genetics and American College of Medical Genetics. Points to consider: ethical, legal and psychosocial implications of genetic testing in children and adolescents. Am J Hum Genet 1995;57:1233. 1006. Green M, Solnit AJ. Reactions to the threatened loss of a child: a vulnerable child syndrome. Pediatrics 1964;034:58. 1007. McIntosh N, Eldrige C. Neonatal death: the neglected side of neonatal care? Arch Dis Child 1984;59:585. 1008. Bourne S. The psychological effects of a stillbirth on women and their doctors. J R Coll Gen Pract 1968;16:103. 1009. Crowther ME. Communication following a stillbirth or neonatal death: room for improvement. Br J Obstet Gynaecol 1995;102:952. 1010. American College of Obstetricians and Gynecologists, Society for Maternal-Fetal Medicine. Management of Stillbirth: Obstetric Care Consensus No, 10. Obstet Gynecol 2020;135:e110. 1011. Causes of death among stillbirths. Stillbirth Collaborative Research Network Writing Group. JAMA 2011;306:2459. 1012. Laury A, Sanchez-Lara PA, Pepkowitz S, et al. A study of 534 fetal pathology cases from prenatal diagnosis referrals analyzed from 1989 through 2000. Am J Med Genet A 2007;143A:3107. 1013. Korteweg FJ, Bouman K, Erwich JJ, et al. Cytogenetic analysis after evaluation of 750 fetal deaths: proposal for diagnostic workup. Obstet Gynecol 2008;111:865. 1014. Saleem S, Tikmani SS, McClure EM, et al. Trends and determinants from stillbirth in developing countries: results from the Global Network’s Population-Based Birth Registry. Reprod Health 2018;15(Suppl 1):100. 1015. Stanley KE, Giordano J, Thorsten V, et al. Casual genetic variants in stillbirth. N Engl J Med 2020;383:1107. 1016. Barrett PM, McCarthy FP, Evans M, et al. Stillbirth is associated with increased risk of long-term maternal renal disease: a nationwide cohort study. Am J Obstet Gynecol 2020;223:427.e1.

1017. Brookes JAS, Hall-Craggs MA, Sams VR, et al. Noninvasive perinatal necropsy by magnetic resonance imaging. Lancet 1996;348:1139. 1018. Gagnon A, Wilson RD, Allen VM. Evaluation of prenatally diagnosed structural congenital anomalies. J Obstet Gynaecol Can 2009;31:875. 1019. Desilets V, Oligny LL. Fetal and perinatal autopsy in prenatally diagnosed fetal abnormalities with normal karyotype. J Obstet Gynaecol Can 2011;267:1047. 1020. Sethi N, Funamoto K, Ingbar C, et al. Noninvasive fetal electrocardiography in the diagnosis of Long QT syndrome: a case series. Fetal Daign Ther 2020;47(9):711. 1021. Shim SH, Ito M, Maher T, et al. Gene sequencing in neonates and infants with the long QT syndrome. Genet Test 2005;9:281. 1022. Cuneo BF, Kaizer AM, Clur SA, et al. Mothers with long QT syndrome are at increased risk for fetal death: findings from a multicenter international study. Am J Obstet Gynecol 2020;222:263.e1. 1023. Lewkowitz AK, Rosenbloom JI, López JD, et al. Association between stillbirth at 23 weeks of gestation or greater and severe maternal morbidity. Obstet Gynecol 2019;134:964. 1024. Nicholas AM, Lewin TJ. Grief reactions of parental couples: congenital handicap and cot death. Med J Aust 1986;144:292. 1025. Lafarge C, Mitchell K, Fox P. Perinatal grief following a termination of pregnancy for foetal abnormality: the impact of coping strategies. Prenat Diagn 2013;33:1173. 1026. Mashiach R, Anter D, Melamed N, et al. Psychological response to multifetal reduction and pregnancy termination due to fetal abnormality. J Matern Fetal Neonatal Med 2013,26:32. 1027. Lewis E, Bryan E. Management of perinatal loss of a twin. BMJ 1988;297:1321. 1028. Lewis E. Stillbirth: psychological consequences and strategies of management. In: Milunsky A, ed. Advances in perinatal medicine, vol. 3. New York: Plenum, 1983:205. 1029. McPhee SJ, Bottles K, Lo B, et al. To redeem them from death: reactions of family members to autopsy. Am J Med 1986;80:665. 1030. Irvin NA, Kennell JH, Klaus MH. Caring for the parents of an infant with a congenital malformation. In: Warkany J, ed. Congenital malformations: notes and comments. Chicago: Year Book Medical Publishers, 1971. 1031. Klaus MH, Kennell JH. Caring for parents of an infant who dies: maternal–infant bonding. St Louis, MO: CV Mosby, 1976.

CHAPTER 1

Genetic Counseling: Preconception, Prenatal, and Perinatal 101

1032. Blood C, Cacciatore J. Parental grief and memento mori photography: narrative, meaning, culture and context. Death Stud 2014;38:224. 1033. Flenady V, Boyle F, Koopmans L, et al. Meeting the needs of parents after a stillbirth or neonatal death. BJOG 2014;121(Suppl 4):137. 1034. Furlong RM, Hobbins JC. Grief in the perinatal period. Obstet Gynecol 1983;61:497. 1035. Shulman LP, Grevengood C, Phillips OP, et al. Family planning decisions after prenatal detection of fetal abnormalities. Am J Obstet Gynecol 1994;171:1373. 1036. Rowe J, Clyman R, Green C, et al. Follow-up of families who experience a perinatal death. Pediatrics 1978;62:166. 1037. Forrest GC, Standish E, Baum JD. Support after perinatal death: a study of support and counseling after bereavement. BMJ 1982;285:1475. 1038. Moldovan R, Pintea S, Austin J. The efficacy of genetic counseling for psychiatric disorders: a meta-analysis. J Genet Couns 2017;26:1341. 1039. Caldwell S, Wusik K, He H, et al. Development and Validation of the Genetic Counseling Self-Efficacy Scale (GCSES). J Genet Couns 2018;27:1248. 1040. Keller H, Wusik K, He H, et al. Further validation of the Genetic Counseling Self-Efficacy Scale (GCSES): its relationship with personality characteristics. J Genet Couns 2020;29(5):748. 1041. Borle K, Morris E, Inglis A, et al. Risk communication in genetic counseling: Exploring uptake and perception of recurrence numbers, and their impact on patient outcomes. Clin Genet 2018;94:239. 1042. Clarke A, Parsons E, Williams A. Outcomes and process in genetic counseling. Clin Genet 1996;50:462. 1043. Voorwinden JS, Plantinga M, Ausems M, et al. Cognitive and affective outcomes of genetic counseling in the Netherlands at group and individual level: a personalized approach seems necessary. Eur J Hum Genet 2020;28:1187. 1044. Montgomery SV, Barsevick AM, Egleston BL, et al. Preparing individuals to communicate genetic test

1045. 1046.

1047. 1048.

1049.

1050.

1051.

1052.

1053.

1054.

1055.

results to their relatives: report of a randomized control trial. Fam Cancer 2013;12:537. Emery AEH, Raeburn JA, Skinner R. Prospective study of genetic counseling. BMJ 1979;1:253. Sibinga MS, Friedman CG. Complexities of parental understanding for phenylketonuria. Pediatrics 1971;48:216. Reynolds BD, Puck MH, Robinson A. Genetic counseling: an appraisal. Clin Genet 1974;5:177. American College of Medical Genetics and Genomics. ACMG policy statement: updated recommendations regarding analysis and reporting of secondary findings in clinical genome-scale sequencing. Genet Med 2015;17:68. Sorenson JR, Swazey JP, Scotch NA. Effective genetic counseling: more informed clients. In: Reproductive pasts, reproductive futures: genetic counseling and its effectiveness. New York: Alan R. Liss, 1981:79. Swerts A. Impact of genetic counseling and prenatal diagnosis for Down syndrome and neural tube defects. Birth Defects Orig Artic Ser 1987;23:61. Kessler S. Psychological aspects of genetic counseling. VI. A critical review of the literature dealing with education and reproduction. Am J Med Genet 1989;34: 340. Davey A, Rostant K, Harrop K, et al. Evaluating genetic counseling: client expectations, psychological adjustment and satisfaction with service. J Genet Couns 2005;14:197. Milunsky A. Your genetic destiny: know your genes, secure your health, save your life. Cambridge, UK: Perseus Books, 2001. Baars MJH, Scherpbier AJJA, Schuwirth LW, et al. Deficient knowledge of genetics relevant for daily practice among medical students nearing graduation. Genet Med 2005;7:295. Marchant G, Barnes M, Evans JP, et al. From genetics to genomics: facing the liability implications in clinical care. J Law Med Ethics 2020;48:11.

2

Preimplantation Genetic Testing Anver Kuliev and Svetlana Rechitsky Herbert Wertheim College of Medicine, Florida International University, Miami, FL and Reproductive Genetic Innovations, Northbrook, IL, USA

Preimplantation genetic testing (PGT1 ) is a practical option for couples at risk of having offspring with serious/fatal chromosomal or monogenic diseases. It has been used for up to 600 monogenic disorders (PGT-M1 ). Moreover, it has been used for human leukocyte antigen (HLA) typing (PGT-HLA1 ), enabling the births of many children whose matched bone marrows have proved life-saving for siblings with congenital and acquired disorders requiring stem cell transplantation treatment. Analysis of single cells or a few cells with a limited amount of available DNA has always presented a technical challenge, especially when PGT is faced with the need for accurate and rapid results from whole-genome amplification (WGA), followed by polymerase chain reaction (PCR) assays that are robust and sensitive. Next-generation sequencing (NGS) has allowed for accurate identification and transfer of euploid embryos (PGT for aneuploidies (PGT-A)1 ). PGT-M was initially applied for the same indications as prenatal diagnosis,2–4 but was then expanded to conditions that had never been considered, such as late-onset diseases with genetic predisposition and preimplantation HLA typing with or without testing for genetic disorders.5–7 PGT represents a natural evolution of the genetic disease prevention technology, from a period with limited genetic counseling and no prenatal diagnosis or treatment to a time when many options, including PGT, have become available.8 Furthermore, PGT has been applied in order to improve

access to the new treatment methods for some severe conditions by stem cell transplantation, for which no traditional treatment approaches are available. The impact of PGT and stem cell treatment on existing policies for the prevention of genetic disease (see Chapter 36) is clear from the increasing use of PGT to avoid unnecessary termination of many wanted pregnancies and for preimplantation HLA typing.

Approaches to preimplantation genetic testing When prenatal genetic diagnosis was first considered in perspective, in 1984, the World Health Organization (WHO) emphasized the relevance of developing earlier approaches for genetic analysis with the possibility of diagnosis before implantation.9, 10 The following possibilities for PGT were mentioned: genetic analysis of the first or second polar bodies and embryo biopsy at the cleavage or blastocyst stage.10, 11 However, these approaches became possible only after introduction of the PCR assay12 and success in micromanipulation and embryo biopsy. First attempts at PGT were undertaken in mammalian embryos over 30 years ago,13–18 when it was demonstrated that cells could be removed from mammalian preimplantation embryos and analyzed successfully without destroying the viability of the embryo in in vitro fertilization (IVF). PGT for human genetic disease was first demonstrated by Handyside et al.19 for X-linked diseases

Genetic Disorders and the Fetus: Diagnosis, Prevention, and Treatment, Eighth Edition. Edited by Aubrey Milunsky and Jeff M. Milunsky. © 2021 Aubrey Milunsky and Jeff M. Milunsky. Published 2021 by John Wiley & Sons Ltd.

102

CHAPTER 2

and by Verlinsky et al.20 for autosomal recessive disorders. Tens of thousands of children without detectable birth defects have been born following these procedures,21–25 demonstrating that PGT can be performed safely in humans. Initially, PGT was based on polar body sampling and embryo biopsy at the cleavage stage, but the present standard shifted to blastocyst biopsy. The polar body approach is still, however, the only possibility for the ethnic groups where no embryos micromanipulation is allowed. The Preimplantation Genetic Diagnosis International Society (PGDIS) and the European Society of Human Reproduction and Embryology (ESHRE) Consortium have published an extensive set of best practice guidelines for PGT.26, 27 These recommendations cover PGT organization, genetic and treatment-related counseling, psychologic evaluation, patient selection, all applicable technical issues, and quality control. The developments of preconception and PGT and the existing problems in the application of these early approaches to clinical practice are presented in this chapter, based on our 30 years’ experience of over 22,000 PGT cycles, including 15,700 PGT-A, 491 PGT-HLA, and 6,778 PGT-M, involving a spectrum of, approximately, 600 different monogenic conditions (Table 2.1). Polar body-based preimplantation genetic testing

The biopsy of gametes opened an intriguing possibility of preconception diagnosis of inherited diseases, because genetic analysis of biopsied gamete material made it realistic to select gametes containing an unaffected allele for fertilization and subsequent transfer.28 In this way, not only was the selective abortion of an affected fetus avoided but also fertilization involving affected gametes, as an option for couples at risk of conceiving a genetically abnormal fetus. Although preconception genetic testing could be achieved by genotyping either oocytes or sperm, the latter approach is still not realistic. Development of methods for culture of primary spermatocytes and spermatogonia followed by genetic analysis of matured spermatides is theoretically possible, but this still remains a subject for future research, such as in the framework of the current attempts at haploidization.29, 30 The technique of sperm duplication has been introduced,

Preimplantation Genetic Testing 103

which may allow testing of the sperm duplicate. However, errors may arise in the reduplication procedure, making the technique of sperm duplication inapplicable for clinical practice.31, 32 The only approach for preconception diagnosis at present, therefore, is genotyping oocytes by biopsy and subsequent genetic analysis of polar bodies. The first attempt to obtain oocyte karyotypes was undertaken in the mouse model by testing the second polar body in the early 1980s, but the technique required much improvement to be considered for clinical application.33 Polar bodies were then used to test the possibility of amplification of β-globin sequences, again in the mouse model.34 The first clinical application of the polar body approach was introduced in 1990.20 It was demonstrated that, in the absence of crossover, the first polar body will be homozygous for the allele not contained in the oocyte and second polar body. However, the first polar body approach will not predict the eventual genotype of the oocytes if crossover occurs, because the primary oocyte in this case will be heterozygous for the abnormal gene. The frequency of crossover varies with the distance between the locus and the centromere, approaching as much as 50 percent for telomeric genes, for which the first polar body approach would be of only limited value, unless the oocytes can be tested further (Figure 2.1). Therefore, the second polar body analysis is required to detect hemizygous normal oocytes resulting after the second meiotic division. In fact, the accumulated experience shows that the most accurate diagnosis can be achieved in cases where the first polar body is heterozygous, so the detection of the normal or mutant gene in the second polar body predicts the opposite mutant or normal genotype of the resulting maternal contribution to the embryo after fertilization.4 To study a possible detrimental effect of the procedure, micromanipulated oocytes were followed and evaluated at different stages of development.3, 4, 35 The absence of any deleterious effect of polar body removal on fertilization, preimplantation, and, possibly, postimplantation development made it possible to consider the polar body approach as a nondestructive test for genotyping the oocytes before fertilization and implantation. In another study, to assess the effect of the second polar body sampling on the viability and

Table 2.1 List of conditions for which preimplantation genetic testing (PGT) was performed and PGT-M outcome: 30 years of original experience. No.

No.

Type of

No.

No.

embryo

embryos

Pregnancy

No.

Conditions

Gene

inheritance

patients

cycles

transfers

transferred

%

deliveries

3-Hydroxyisobutyryl-CoA hydrolase deficiency (HIBCHD)

HIBCH

AR

1

1

1

2

0

0

3-Methylglutaconic aciduria with deafness, encephalopathy, and

SERAC1

AR

1

1

1

1

0

0

Achondroplasia (ACH)

FGFR3

AD

7

6

Achromatopsia 2 (ACHM2)

CNGA3

AR

1

1

1

1

1

1

Achromatopsia 3 (ACHM3)

CNGB3

AR

3

4

4

5

2

2

Leigh-like syndrome (MEGDEL)

Acromesomelic dysplasia, Maroteaux type (AMDM)

NPR2

AR

8

1

17

1

11

2

14

2

1

1

Acyl-CoA dehydrogenase, medium-chain, deficiency

ACADM

AR

3

8

7

14

4

4

Acyl-CoA dehydrogenase, very long-chain; (ACADVL)

ACADVL

AR

5

6

6

11

2

2

Adrenal hyperplasia, congenital, due to 21-hydroxylase deficiency

CYP21A2

AR

23

34

26

42

17

17

Adrenoleukodystrophy (ALD)

ABCD1

XL

17

33

20

29

11

11

Agammaglobulinemia, X-linked (XLA)

BTK

XL

4

7

7

13

3

3

AR

1

2

2

2

1

Alagille syndrome 1 (ALGS1)

JAG1

AD

1

1

1

1

1

1

Albinism, ocular, type i (OA1)

Aicardi–Goutieres syndrome 5 (AGS5 + CF)

GPR143

XL

1

12

5

9

4

3

Albinism, oculocutaneous, type ia (OCA1a)

TYR

AR

4

7

6

9

3

3

Albinism, oculocutaneous, type ii (OCA2)

OCA2

AR

3

6

5

9

3

3

Albinism, oculocutaneous, type iii (OCA3)

TYRP1

AR

1

1

0

0

0

0

Allan–Herndon–Dudley syndrome (AHDS)

SLC16A2

XL

1

2

2

2

1

1

Alopecia universalis congenita (ALUNC)

HR

AR

1

1

1

2

1

1

Alpha-1-antitrypsin deficiency (A1ATD)

SERPINA1

AR

9

16

14

18

9

8

Alport syndrome, autosomal dominant

COL4A3

AR

1

4

0

0

0

0

Alport syndrome, X-linked (ATS)

SAMHD1

8

16

1

COL4A5

XL

15

22

10

9

Alzheimer disease 3

PSEN1

AD

2

3

3

6

3

3

Alzheimer disease 4

PSEN2

AD

1

1

1

2

0

0

Alzheimer disease (AD)

APP

AD

2

3

2

4

2

1

Amegakaryocytic thrombocytopenia, congenital (CAMT)

MPL

AR

1

1

0

0

0

Amyloidosis, hereditary, transthyretin-related

TTR

AD

3

7

5

6

3

2

Amyotrophic lateral sclerosis 1 (ALS1)

SOD1

XL

2

2

2

3

2

1

Amyotrophic lateral sclerosis 4, juvenile (ALS4)

SETX

AD

1

1

1

1

1

0

1

Anemia, nonspherocytic hemolytic, due to g6pd deficiency

G6PD

XL

9

12

12

15

6

6

Angelman syndrome (AS)

UBE3A

AD

2

2

2

3

1

1

Angioedema, hereditary, type i (HAE1)

C1NH

AD

3

4

3

4

1

1

Aniridia (AN)

PAX6

AD

4

7

5

6

4

4

NOTCH1

AD

1

1

2

2

1

1

Argininosuccinic aciduria

ASL

AR

2

3

3

4

1

1

Arterial tortuosity syndrome (ATS)

Aortic valve disease 1 (AOVD1)

SLC2A10

AR

1

2

2

2

1

1

Arthrogryposis, distal, type 2a (DA2a)

MYH3

AD

1

2

2

2

1

1

Arthrogryposis, distal, type 2b (DA2b)

TNNI2

AD

1

2

1

1

0

0

Arthrogryposis, distal, type 2b (DA2b)

TNNT3

AD

1

3

2

3

2

1

Arthrogryposis, distal, type 9 (DA9)

FBN2

AD

1

2

2

2

2

2

Ataxia-telangiectasia (AT)

ATM

AD

5

12

7

8

6

5

Auriculocondylar syndrome 2 (ARCND2)

PLCB4

AR

1

1

0

0

0

0

Axenfeld–rieger syndrome, type 1 (RIEG1)

PITX2

AD

3

13

13

15

5

4

Bardet–Biedl syndrome 10 (BBS10)

BBS10

AR

1

2

3

4

1

1

Bardet–Biedl syndrome 2 (BBS2)

BBS2

AR

1

1

2

2

2

1

BBS4

AR

1

1

2

2

1

1

Bartter syndrome, type 3 (BARTS3)

CLCNKB

AR

1

1

2

2

1

1

Basal cell nevus syndrome (BCNS) (Gorlin)

Bardet–Biedl syndrome 4 (BBS4)

PTCH1

AD

6

7

6

10

4

4

Benign chronic pemphigus (BCPM)

ATP2C1

AD

1

1

1

1

1

0

Beta-ureidopropionase deficiency (UPB1D)

UPB1

AR

1

1

2

2

2

1

Biotinidase deficiency

BTD

AR

3

5

2

3

2

2

Birt–Hogg–Dube syndrome (BHD)

FLCN

AD

1

2

1

1

1

1

Bleeding disorder, platelet-type, 16 (BDPLT16)

ITGB3

AD

1

1

0

0

0

0

Blepharophimosis, ptosis, and epicanthus inversus (BPES)

FOXL2

AD

3

7

5

7

3

3

Blood group – Kell–Cellano system

KEL

AR

14

32

19

32

5

5

Brachydactyly, type B1 (BDB1)

ROR2

AD

1

3

2

4

2

2

Branchiooculofacial syndrome (BOFS)

TFAP2A

AD

1

1

1

2

0

0

Breast cancer

PALB2

AD

2

4

2

2

1

1

Breast–ovarian cancer, familial, susceptibility to, 1 (BROVCA1)

BRCA1

AD

93

175

128

183

89

83

87

122

55

51

Campomelic dysplasia with autosomal sex reversal

SOX9

AD

1

1

0

0

0

0

Camurati–Engelmann disease (CAEND)

Breast–ovarian cancer, familial, susceptibility to, 2 (BROVCA2)

TGFB1

BRCA2

AD

AD

1

64

1

123

1

1

0

0

Canavan disease

ASPA

AR

4

6

5

7

5

5

Carbamoyl phosphate synthetase i deficiency

CPS1

AR

1

1

1

2

0

0

Cardioencephalomyopathy, fatal infantile, due to cytochrome c oxidase

SCO2

AR

2

5

5

10

3

3

Cardiomyopathy, dilated, 1A (CMD1A)

LMNA

AR

7

17

16

25

10

8

Cardiomyopathy, dilated, 1DD (CMD1DD)

RBM20

AD

1

2

2

2

2

2

deficiency 1

(Continued)

Table 2.1 (Continued) No.

No.

Type of

No.

No.

embryo

embryos

Pregnancy

No.

Conditions

Gene

inheritance

patients

cycles

transfers

transferred

%

deliveries

Cardiomyopathy, dilated, 1E (CMD1E)

SCN5A

AD

1

2

2

2

1

1

Cardiomyopathy, dilated, 1G (CMD1G)

TTN

AD

2

2

3

3

1

1

Cardiomyopathy, dilated, 1S (CMD1S)

MYH7

AD

3

6

4

4

2

2

Cardiomyopathy, dilated, with woolly hair, keratoderma, and tooth

DSP

AD

2

3

2

3

2

1

TNNT2

AD

1

2

1

1

1

agenesis (DCWHKTA) Cardiomyopathy, familial hypertrophic, 2 (CMH2)

0

Cardiomyopathy, familial hypertrophic, 4 (CMH4)

MYBPC3

AD

14

22

16

23

11

9

Cardiomyopathy, familial hypertrophic, 7 (CMH7)

TNNI3

AD

1

1

1

1

0

0

Cardiomyopathy, familial hypertrophic, 8 (CMH8)

MYL3

AD

1

2

0

0

0

0

Carnitine deficiency, systemic primary (CDSP)

SLC22A5

AR

1

2

1

2

1

1

Carnitine palmitoyltransferase II deficiency, infantile

CPT2

AR

4

7

4

4

2

2

Cerebral arteriopathy, autosomal dominant

NOTCH3

AD

3

7

6

6

6

4

Cerebral creatine deficiency syndrome 1 (CCDS1)

SLC6A8

XL

1

1

1

2

1

1

Ceroid lipofuscinosis, neuronal 2, late infantile (CLN2)

TPP1

AR

2

3

2

2

2

1

Ceroid lipofuscinosis, neuronal, 10 (CLN10)

CTSD

AR

1

1

2

3

1

1

Ceroid lipofuscinosis, neuronal, 5 (CLN5)

CLN5

AR

1

1

2

3

0

0

Ceroid lipofuscinosis, neuronal, 6 (CLN6)

CLN6

AR

2

2

1

2

0

0

Charcot–Marie–Tooth disease, axonal, type 2A2 (CMT2A2)

MFN2

AD

2

9

6

7

2

Charcot–Marie–Tooth disease, axonal, type 2B (CMT2B)

RAB7A

AD

1

1

2

4

2

1

Charcot–Marie–Tooth disease, axonal, type 2E (CMT2E)

NEFL

AD

1

4

4

7

1

1 0

Charcot–Marie–Tooth disease, axonal, type 2F (CMT2F)

HSPB1

AD

1

1

1

1

0

Charcot–Marie–Tooth disease, demyelinating, type 1A (CMT1A)

PMP22

AD

28

56

38

51

25

2

21

Charcot–Marie–Tooth disease, demyelinating, type 1B (CMT1B)

MPZ

AD

2

5

2

5

0

0

Charcot–Marie–Tooth disease, X-linked, 1 (CMTX1)

GJB1

XL

6

9

9

14

5

5

Cholestasis, benign recurrent intrahepatic, 2 (BRIC2)

ABCB11

AR

1

2

2

4

1

1

Cholestasis, progressive familial intrahepatic, 3 (PFIC3)

ABCB4

AR

1

1

1

2

1

1

Chondrodysplasia punctata 1, X-linked recessive (CDPX1)

ARSE

XL

1

2

2

3

0

0

XL

3

5

5

9

Ciliary dyskinesia, primary, 15 (CILD15)

CCDC40

AR

1

1

1

1

1

1

Ciliary dyskinesia, primary, 3 (CILD3)

Choroideremia (CHM)

DNAH5

CHM

AR

2

2

1

2

1

3

1

3

Citrullinemia, classic

ASS1

AR

4

7

6

8

3

3

Cleidocranial dysplasia (CCD)

RUNX2

AD

1

3

5

5

Cockayne syndrome A (CSA)

ERCC8

AR

1

1

2

2

1

1

Coenzyme Q10 deficiency, primary, 7 (COQ10D7)

COQ4

AR

1

1

1

1

1

2

1

2

Cohen syndrome (COH1)

VPS13B

AR

2

2

2

4

2

2

Colorectal cancer, hereditary nonpolyposis, type 1 (HNPCC1)

MSH2

AD

11

21

14

17

7

6

Colorectal cancer, hereditary nonpolyposis, type 2 (HNPCC2)

MLH1

AD

10

18

15

25

9

Colorectal cancer, hereditary nonpolyposis, type 4 (HNPCC4)

PMS2

AD

1

2

1

1

0

0

Colorectal cancer, hereditary nonpolyposis, type 5 (HNPCC5)

MSH6

AD

5

10

8

11

5

5

Combined oxidative phosphorylation deficiency 13 (COXPD13)

PNPT1

AR

1

1

3

5

0

0

Cone–rod dystrophy 6 (CORD6)

GUCY2D

AD

1

1

1

0

0

9

0

Congenital disorder of deglycosylation (CDDG)

NGLY1

AR

1

1

2

2

1

1

Congenital disorder of glycosylation, type Ia (CDG1A)

PMM2

AR

5

5

4

4

3

3

Congenital disorder of glycosylation, type IIc (CDG2C)

SLC35C1

AR

1

1

2

3

0

0

Congenital disorder of glycosylation, type IIL (CDG2L)

COG6

AR

1

2

2

2

0

0

Congenital disorder of glycosylation, type In (CDG1N)

RFT1

AR

2

2

2

4

1

1 1

Cranioectodermal dysplasia 2 (CED2)

WDR35

AR

1

1

1

1

1

Craniofrontonasal syndrome (CFNS)

EFNB1

XL

1

1

1

1

0

Creutzfeldt–Jakob disease (CJD); Gerstmann–Straussler disease (GSD)

PRNP

AD

6

9

9

12

8

7

Crouzon syndrome

FGFR2

AD

8

16

14

23

9

8

Currarino syndrome

MNX1

AD

1

1

1

2

1

1

Cutis laxa, autosomal dominant 1 (ADCL1)

ELN

AD

2

2

Cutis laxa, autosomal recessive, type IIB (ARCL2B)

PYCR1

AR

1

1

2

2

1

1

Cutis laxa, autosomal recessive, type IIIA (ARCL3A)

ALDH18A1

AR

1

1

1

1

1

1

Cystic fibrosis (CF)

CFTR

AR

1

496

4

748

3

627

4

0

1072

354

314

Cystinosis, nephropathic (CTNS)

CTNS

AR

1

1

1

1

0

0

Danon disease

LAMP2

XL

1

2

2

2

2

2

Darier–White disease (DAR)

ATP2A2

AD

1

1

1

1

1

1

D-bifunctional protein deficiency

HSD17B4

AR

1

1

1

1

0

0

Deafness, autosomal dominant 3b (DFNA3b)

GJB6

AD

1

2

2

3

1

1

Deafness, neurosensory, autosomal recessive 1 (DFNB1)

GJB2

AR

51

68

56

80

33

30

Dentinogenesis imperfecta, shields type III

DSPP

AD

1

2

2

2

2

1

Developmental delay

DHX35

AR

1

1

2

2

1

1

Diabetes insipidus, nephrogenic, X-linked

AVPR2

XL

1

3

3

3

1

1

Diabetes mellitus, permanent neonatal (PNDM)

INS

AD

1

1

1

1

1

1 (Continued)

Table 2.1 (Continued) No.

No.

Type of

No.

No.

embryo

embryos

Pregnancy

No.

Conditions

Gene

inheritance

patients

cycles

transfers

transferred

%

deliveries

Diamond–Blackfan anemia 1 (DBA1)

RPS19

AD

1

1

1

2

1

1

Digeorge syndrome (DGS)

TBX1

AD

1

1

1

1

1

Dihydrolipoamide dehydrogenase deficiency (DLDD)

DLD

AR

1

1

1

1

1

1

Donnai–Barrow syndrome

LRP2

AR

1

1

0

0

0

0

Dyskeratosis congenita, autosomal dominant 3 (DKCA3)

TINF2

AD

1

2

2

3

1

1

1

Dyskeratosis congenita, autosomal dominant 2 (DKCA2)

TERT

AD

1

3

1

1

0

Dyskeratosis congenita, autosomal recessive 5 (DKCB5)

RTEL1

AR

1

1

2

2

1

1

Dyskeratosis congenita, X-linked (DKCX)

DKC1

XL

1

1

1

2

1

1

Dyskinesia, seizures, and intellectual developmental disorder (DYSEIDD)

DEAF1

AR

1

1

1

1

0

0

0

Dystonia 1, torsion, autosomal dominant (DYT1)

TOR1A

AD

16

36

35

63

18

18

Dystonia 28, childhood-onset (DYT28)

KMT2B

AD

1

1

1

1

0

0

Dystonia 3, torsion, X-linked (DYT3)

TAF1

XL

1

1

1

2

1

1

Ectodermal dysplasia 10b, hypohidrotic/hair/tooth type, autosomal

EDAR

AR

1

1

1

2

1

1

recessive (ECTD10B) Ectodermal dysplasia, hypohidrotic, X-linked (XHED)

EDA

XL

6

8

8

10

4

4

Ehlers–Danlos syndrome, classic type

COL5A1

AD

2

4

3

4

3

2

Ehlers–Danlos syndrome, type IV, autosomal dominant

COL3A1

AD

4

6

4

7

4

3

Ehlers–Danlos syndrome, type VI (EDS6)

PLOD1

AR

1

1

2

3

0

0

Emery–Dreifuss muscular dystrophy 1, X-linked (EDMD1)

EMD

XL

3

4

4

7

3

Epidermolysis bullosa dystrophica, autosomal dominant (DDEB)

COL7A1

AR

8

9

8

13

4

4

Epidermolysis bullosa simplex with pyloric atresia (EBSPA)

PLEC1

AR

1

2

1

3

1

1

Epidermolysis bullosa simplex, dowling-meara type (EBSDM)

KRT5

AD

1

2

1

2

1

3

1

Epidermolysis bullosa, junctional, Herlitz type

LAMA3

AR

4

9

7

13

7

7

Epidermolysis bullosa, junctional, non-herlitz type

LAMB3

AR

5

6

5

9

2

2

Epidermolytic hyperkeratosis (EHK)

KRT10

AD

2

3

2

2

2

2

Epileptic encephalopathy, early infantile, 2 (EIEE2)

CDKL5

XL

1

1

1

2

1

Epileptic encephalopathy, early infantile, 3 (EIEE3)

SLC25A22

AR

1

1

0

0

0

0

Epileptic encephalopathy, early infantile, 5 (EIEE5)

SPTAN1

AR

1

1

0

0

0

0

1

Epiphyseal dysplasia, multiple, 1 (EDM1)

COMP

AD

3

4

2

2

1

1

Exostoses, multiple, type I

EXT1

AD

11

21

17

29

12

Exostoses, multiple, type II

EXT2

AD

3

8

6

10

3

3

Fabry disease

GLA

XL

12

19

14

22

9

7

10

Facioscapulohumeral muscular dystrophy 1 (FSHD1)

FRG1

AD

25

51

42

71

23

20

Factor VII deficiency

F7

AR

1

1

1

1

0

0

Familial adenomatous polyposis 1 (FAP1)

APC

AD

23

44

36

57

17

15

Familial cold autoinflammatory syndrome 1 (FCAS1)

NLPR3

AD

1

1

1

Familial Mediterranean fever (FMF)

MEFV

AR

10

18

16

22

11

8

Fanconi anemia, complementation group A (FANCA)

FANCA

AR

2

5

2

3

2

2

Fanconi anemia, complementation group C (FANCC)

FANCC

AR

2

5

4

8

1

1

Fetal akinesia deformation sequence (FADS)

NUP88

1

1

1

AR

1

1

1

2

1

1

Fetal akinesia deformation sequence (FADS)

RAPSN

AR

1

1

1

2

1

0

Fragile-X mental retardation syndrome

FMR1

XL

312

608

450

662

243

214

Fraser syndrome 1 (FRASRS1)

FRAS1

AR

2

2

2

2

1

1

Friedreich ataxia 1 (FRDA)

FXN

AR

2

6

4

7

2

2

Frontotemporal dementia and/or amyotrophic lateral sclerosis 1

c9orf72

AD

1

1

1

1

1

1

ALDOB

AR

2

7

6

7

3

(FTDALS1) Fructose intolerance, hereditary

3

Fumarase deficiency (FMRD)

FH

AR

1

1

0

0

0

0

Galactosemia

GALT

AR

3

7

5

6

2

2

Gastric cancer, hereditary diffuse (HDGC)

CDH1

AD

1

1

1

2

Gaucher disease, type I

GBA

XL

39

52

34

57

24

19

Geroderma osteodysplasticum (GO)

GORAB

AR

1

2

2

4

1

1

1

1

Gitelman syndrome (GTLMNS)

SLC12A3

AR

1

1

1

1

0

0

Glaucoma 3, primary congenital, A (GLC3A)

CYP1B1

AR

1

1

2

2

1

1

Glut1 deficiency syndrome 1 (GLUT1DS1)

SLC2A1

AD

1

2

1

2

0

0

Glutaric acidemia I

GCDH

AR

1

1

1

2

0

0

Glycine encephalopathy (GCE)

GLDC

AR

6

7

6

11

6

6

Glycogen storage disease Ia (GSD1A)

G6PC

AR

1

1

2

2

0

Glycogen storage disease II (GSD2)

GAA

AR

5

7

4

9

1

1

Glycogen storage disease IXa1 (GSD9A1)

PHKA2

XL

1

1

0

0

0

0

Glycogen storage disease VII (GSD7)

PFKM

AR

1

1

1

1

1

1

Gm1-gangliosidosis, type I

GLB1

AR

5

5

5

10

4

0

4

Granulomatous disease, chronic, X-linked (CDGX)

CYBB

XL

4

5

4

6

3

2

Greig cephalopolysyndactyly syndrome (GCPS)

GLI3

AD

1

1

2

2

0

0

Harel–Yoon syndrome (HAYOS)

ATAD3A

AR

1

3

3

3

1

1

Hemoglobin-alpha locus 1 (HBA1)

HBA

AR

14

23

21

38

10

10

470

402

762

192

161

1

0

0

0

0

Hemoglobin-beta locus (HBB)

HBB

AR

301

Hemophagocytic lymphohistiocytosis, familial, 2 (FHL2)

PRF1

AR

1

(Continued)

Table 2.1 (Continued) No.

No.

Type of

No.

No.

embryo

embryos

Pregnancy

No.

Conditions

Gene

inheritance

patients

cycles

transfers

transferred

%

deliveries

Hemophagocytic lymphohistiocytosis, familial, 3 (FHL3)

UNC13D

AR

3

4

4

5

4

Hemophilia A (HEMA)

F8

XL

62

103

88

145

50

42

Hemophilia B (HEMB)

F9

XL

5

6

6

9

6

6

Hereditary leiomyomatosis and renal cell cancer (HLRCC)

FH

AD

1

1

1

2

0

3

0

Hereditary motor and sensory neuropathy, type IIC (HMSN2C)

TRPV4

AD

1

1

2

2

1

1

Hermansky–Pudlak syndrome 1 (HPS1)

HPS1

AR

1

4

3

6

2

2

HLA + myelodysplastic syndrome (MDS)

GATA2

AD

1

2

1

1

1

1 2

HLA + Shwachman–Diamond syndrome (SDS)

SBDS

AR

4

10

3

4

2

HLA + adenosine deaminase deficiency (ADA)

ADA

AR

1

1

1

1

1

1

HLA + adrenoleukodystrophy

ABCD1

XL

3

7

2

2

1

1

HLA + Diamond–Blackfan anemia 1 (DBA1)

RPS19,

AD

6

13

10

15

5

5

HLA + Diamond–Blackfan anemia 2 (DBA2)

RPS20

AD

1

1

1

1

1

1

HLA + Diamond–Blackfan anemia 3 (DBA3)

RPS 24

AD

1

1

1

1

0

0

HLA + Diamond–Blackfan anemia 5 (DBA5)

RPL35A

AD

1

1

1

1

1

1

HLA + Diamond–Blackfan anemia 9 (DBA9)

RPS10

AD

1

1

2

2

1

1

HLA + ectodermal dysplasia, hypohidrotic, with immune deficiency

IKBKG

XL

2

9

6

8

2

2

HLA + Fanconi anemia, complementation group A (FANCA)

FANCA

AR

18

52

29

43

14

10

HLA + Fanconi anemia, complementation group C (FANCC)

FANCC

AR

2

5

5

8

1

1

HLA + Fanconi anemia, complementation group D2 (FANCD2)

FANCD2

AR

1

3

2

3

1

1

HLA + Fanconi anemia, complementation group F (FANCF)

FANCF

AR

2

5

2

3

0

0

HLA + Fanconi anemia, complementation group G (FANCG)

FANCG

AR

2

2

2

3

2

2

HLA + Fanconi anemia, complementation group I (FANCI)

FANCI

AR

1

2

2

3

0

0

HLA + Fanconi anemia, complementation group J (FANCJ)

BRIP1

AR

1

1

1

1

1

1

HLA + Fanconi anemia, complementation group JI (FANCJ)

BRIP1

AR

1

3

1

3

0

0

1

2

2

4

1

0

AR

1

3

2

2

1

1 6

HLA + Glanzmann thrombasthenia (GT, +DMD)

ITGA2B DMD

HLA + granulomatous disease, chronic, autosomal recessive, cytochrome

NCF1

b-positive, type I (CDG1) HLA + granulomatous disease, chronic, X-linked (CDGX)

CYBB

XL

6

16

13

17

7

HLA + hemoglobin-beta locus (HBB)

HBB

AR

92

188

119

177

41

HLA + hyper-IgE recurrent infection syndrome, autosomal recessive

DOCK8

AR

1

1

0

0

0

0

HLA + Krabbe disease

GALC

AR

1

1

1

2

1

1

HLA + myotonic dystrophy 1 (DM1)

DMPK

AD

1

2

1

2

1

1

HLA + neutropenia, severe congenital, 1, autosomal dominant (SCN1)

ELANE

AD

2

3

2

5

2

1

31

HLA + polycystic kidney disease 1 (PKD1)

PKD1

AD

1

1

1

2

1

1

HLA + sickle cell anemia

HBB

AR

18

29

18

27

12

8

HLA + thrombocythemia 1 (THCYT1 )

SH2B3

AR

1

2

2

2

2

1

HLA + thrombotic thrombocytopenic purpura, congenital (TTP)

ADAMTS13

AR

1

2

2

4

1

1

HLA + Wiskott–Aldrich syndrome (WAS)

WAS

XL

1

1

0

0

0

0

HLA immunodeficiency with hyper-IgM, type 1 (HIGM1)

CD40LG

XL

8

15

9

13

5

4

HLA + pyruvate kinase deficiency of red cells

PKLR

AD

1

2

1

1

0

0

Holoprosencephaly 2 (HPE2)

SIX3

AD

1

1

1

2

0

0

Holt–Oram syndrome (HOS)

TBX5

AD

5

8

8

9

4

4

Homocystinuria due to cystathionine beta-synthase deficiency

CBS

AR

4

6

4

9

3

3

Homocystinuria due to deficiency of n(5,10)-methylenetetra-

MTHFR

AR

1

1

1

2

0

0 0

hydrofolate reductase activity MTR

AR

1

2

1

1

0

Human leukocyte antigens

Homocystinuria-megaloblastic anemia, cblG complementation type

HLA

AR

60

119

73

108

25

Huntington disease (HD)

HTT

AD

141

209

171

267

107

97

Hurler syndrome

IDUA

AR

7

10

8

13

3

3

Hyaline fibromatosis syndrome (HFS)

ANTXR2

AR

1

1

1

2

1

1

(HMAG) 20

Hydrocephalus due to congenital stenosis of aqueduct of Sylvius (HSAS)

L1CAM

XL

11

16

16

34

8

6

Hydroxyacyl-CoA dehydrogenase/3-ketoacyl-CoA thiolase/enoyl-CoA

HADHA

AR

4

4

4

13

3

3

hydratase, alpha subunit (HADHA) Hyperinsulinemic hypoglycemia, familial, 1 (HHF1)

ABCC8

AR

2

11

8

19

4

2

Hyperuricemic nephropathy, familial juvenile, 1 (HNFJ1)

UMOD

AD

1

1

1

1

0

0

ANOS1

XL

1

1

2

2

0

0

Hypogonadotropic hypogonadism 1 with or without anosmia (HH1)

KAL1

XL

1

2

1

1

1

1

Hypoparathyroidism–retardation–dysmorphism syndrome (HRDS)

Hypogonadotropic hypogonadism 1 with or without anosmia (HH1)

TBCE

1R

1

1

1

2

0

0

Hypophosphatasia, infantile

ALPL

AR

6

7

6

9

4

4

Ichthyosis, congenital, autosomal recessive 1 (ARCI1)

TGM1

AD

2

9

7

10

1

1

Ichthyosis, lamellar, 2 (LI2)

ABCA12

AR

2

2

1

2

0

0

ELOVL4

AR

1

1

1

1

0

0

Ichthyosis, X-linked (XLI)

STS

XL

2

3

3

4

1

1

Ifap syndrome with or without Bresheck syndrome

Ichthyosis, spastic quadriplegia, and mental retardation (ISQMR)

MBTPS2

XL

2

3

2

5

2

1

Immunodeficiency with hyper-IgM, type 1 (HIGM1)

CD40LG

XL

4

14

14

22

6

6

Immunodysregulation, polyendocrinopathy, and enteropathy, X-linked

FOXP3

XL

2

3

3

3

1

1

IKBKG

XL

15

35

28

43

11

(IPEX) Incontinentia pigmenti (IP)

11 (Continued)

Table 2.1 (Continued) No.

No.

Type of

No.

No.

embryo

embryos

Pregnancy

No.

Conditions

Gene

inheritance

patients

cycles

transfers

transferred

%

deliveries

Infantile cerebellar-retinal degeneration (ICRD)

ACO2

AR

1

1

1

2

2

2

Infantile liver failure syndrome 1 (ILFS1)

LARS

AR

1

1

2

2

1

1

Isovaleric acidemia (IVA)

IVD

AR

1

1

1

2

0

0

Joubert syndrome 1 (JBTS1)

INPP5E

AR

1

1

2

2

1

1

Joubert syndrome 17 (JBTS17)

CPLANE1

AD

1

1

1

2

1

1

Joubert syndrome 2 (JBTS2)

TMEM216

AR

1

1

2

2

1

1

Joubert syndrome 21 (JBTS21)

CSPP1

AR

2

5

4

7

1

1

Joubert syndrome 23 (JBTS23)

KIAA0586

AR

1

1

1

2

1

1 0

AR

1

0

0

Joubert syndrome 6 (JBTS6)

TMEM67

AR

2

3

2

2

2

Krabbe disease

Joubert syndrome 3 (JBTS3)

GALC

AR

11

12

11

19

7

5

Larsen syndrome (LRS)

FLNB

AD

2

2

1

1

1

1

Leber congenital amaurosis 2 (LCA2)

RPE65

AR

1

1

0

0

0

0

Leigh syndrome (LS)

AHI1

NDUFS8

1

0

2

AR

1

1

0

0

0

Leigh syndrome (LS)

SURF1

AR

1

1

1

3

0

0

Lesch–Nyhan syndrome (LNS)

HPRT1

XL

1

4

3

3

2

2

0

Leukoencephalopathy with vanishing white matter (VWM)

EIF2B2

AR

1

1

1

2

1

0

Li–Fraumeni syndrome 1 (LFS1)

TP53

AD

16

22

17

24

13

11

Lipoid congenital adrenal hyperplasia (LCAH)

STAR

AR

1

2

2

3

1

1

ARX

XL

0

0

Loeys–Dietz syndrome 1 (LDS1)

TGFBR2

AD

2

5

4

6

2

1

Long QT syndrome 1 (LQT1)

Lissencephaly, X-linked, 2 (LISX2)

KCNQ1

AD

4

5

2

2

2

2

Long QT syndrome 2 (LQT2)

KCNH2

1

1

1

2

AD

3

3

2

2

1

Long QT syndrome 8 (LQT8)

CACNA1C

AD

1

1

1

1

1

1

Lymphedema, hereditary, III (LMPH3)

PIEZO1

AR

1

1

0

0

0

0

1

Lymphoproliferative syndrome, X-linked, 1 (XLP1)

SH2D1A

XL

1

1

2

3

2

2

Lysosomal acid lipase deficiency

LIPA

AR

2

2

2

4

2

2

Machado–Joseph disease (MJD)

ATXN3

AD

4

7

6

8

6

6

Macular dystrophy, vitelliform, 2 (VMD2)

BEST1

AD

1

1

Maple syrup urine disease (MSUD)

BCKDHB

AR

1

2

2

2

1

1

Marfan syndrome (MFS)

FBN1

AD

30

1

58

1

46

1

78

1

27

21

Marinesco–Sjogren syndrome (MSS)

SIL1

AR

1

3

3

5

1

1

Meckel syndrome, type 1 (MKS1)

MKS1

AR

2

Meckel syndrome, type 4 (MKS4)

CEP290

4

6

6

10

4

Meckel syndrome, type 6 (MKS6)

CC2D2A

AR

2

5

5

9

2

2

Meckel syndrome, type 6 (MKS6)

CCD2DA2

AR

1

1

1

2

1

1

Meckel syndrome, type 8 (MKS8)

TCTN2

AR

1

5

1

5

2

9

2

2

1

2 4

1

Mental retardation, autosomal dominant 35 (MRD35)

PPP2R5

AD

1

1

2

2

1

1

Mental retardation, autosomal recessive 38 (MRT38)

HERC2

AR

1

2

2

3

1

1

Metachromatic leukodystrophy due to saposin B deficiency

PSAP

AR

1

1

0

0

0

0

Metachromatic leukodystrophy (MLD)

ARSA

AR

3

4

3

4

4

2

Metaphyseal chondrodysplasia, Schmid type (MCDS)

COL10A1

AD

2

7

3

4

2

2

Methylmalonic aciduria and homocystinuria, cblC type

MMACHC

AR

3

6

6

11

5

5

Methylmalonic aciduria due to methylmalonyl-CoA mutase deficiency

MUT

AR

2

4

4

4

2

2

Methylmalonic aciduria, cblB type

MMAB

AR

3

3

2

3

1

1

Microcephalic osteodysplastic primordial dwarfism, type I (MOPD1)

RNU4ATAC

AR

1

1

1

1

1

1

Microcephaly 2, primary, autosomal recessive (MCPH2)

WDR62

AR

1

1

1

1

0

0

Microcephaly 5, primary, autosomal recessive (MCPH5)

ASPM

AR

2

3

2

3

2

2

Microcephaly 6, primary, MCPH6)

CENPJ

AR

1

2

2

2

1

1

Microphthalmia, isolated, with coloboma 3 (MCOPCB3)

VSX2

AR

2

2

1

1

1

1

Midface hypoplasia, hearing impairment, elliptocytosis, and

AMMECR1

XL

2

8

6

9

2

2

nephrocalcinosis (MFHIEN) Migraine, familial hemiplegic, 1 (FHM1)

CACNA1A

AD

1

1

1

2

1

1

Mitochondrial complex i deficiency due to acad 9 deficiency

ACAD9

AR

1

1

1

2

1

1

Mitochondrial DNA depletion syndrome 13

FBXL4

AD

1

1

3

4

1

1

Mitochondrial DNA depletion syndrome 4a (Alpers type) (MTDPS4A)

POLG

AR

3

5

5

5

4

4

Molybdenum cofactor deficiency, complementation group B (MOCODB)

MOCS2

AR

1

1

3

4

0

0

Mosaic variegated aneuploidy syndrome 1 (MVA1)

BUB1B

AR

1

1

1

2

1

0

Mucolipidosis II alpha/beta

GNPTAB

AR

2

3

Mucopolysaccharidosis, type II (MPS2)

IDS

XL

9

20

15

29

10

6

Mucopolysaccharidosis, type IIIA (MPS3A)

SGSH

AR

2

2

2

2

3

2

0

2

0

2

Mucopolysaccharidosis, type IVA (MPS4A)

GALNS

AR

1

4

4

12

2

2

Multinucleated neurons, anhydramnios, renal dysplasia, cerebellar

1

CEP55

AR

1

1

1

2

1

Multiple congenital anomalies–hypotonia–seizures syndrome 2 (MCAHS2)

PIGA

XL

1

1

0

0

0

0

Multiple endocrine neoplasia, type I (MEN1)

MEN1

AD

8

21

16

23

7

4

hypoplasia

Multiple endocrine neoplasia, type IIA (MEN2A)

RET

AD

6

11

11

17

8

Multiple endocrine neoplasia, type IV (MEN4)

CDKN1B

AD

1

3

1

1

1

8 1 (Continued)

Table 2.1 (Continued) No.

No.

Type of

No.

No.

embryo

embryos

Pregnancy

No.

Conditions

Gene

inheritance

patients

cycles

transfers

transferred

%

deliveries

Muscular dystrophy, congenital merosin-deficient, 1A (MDC1A)

LAMA2

AR

6

7

7

14

7

Muscular dystrophy, Duchenne type (DMD)

DMD

XL

69

115

103

169

57

48

Muscular dystrophy, limb-girdle, type 2A (LGMD2A)

CAPN3

AR

1

1

0

0

0

0

6

Muscular dystrophy, limb-girdle, type 2S (LGMD2S)

TRAPPC11

AR

1

1

2

2

2

2

Muscular dystrophy–dystroglycanopathy (congenital with brain and eye

FKRP

AR

1

3

3

3

1

1 2

anomalies), type A, 5 (MDDGA5) Muscular dystrophy–dystroglycanopathy (congenital with brain and eye

FKTN

AR

2

2

2

3

2

Myoglobinuria, acute recurrent, autosomal recessive

LPIN1

AR

1

1

1

1

1

1

Myopathy, areflexia, respiratory distress, and dysphagia, early-onset

MEGF10

AR

1

1

1

1

1

1

anomalies), type A, 4 (MDDGA4)

(EMARDD) Myopathy, centronuclear, X-linked (CNMX)

MTM1

XL

5

6

4

6

4

4

Myopathy, myofibrillar, 1 (MFM1)

DES

AD

1

1

1

1

1

1

Myotonia congenita, autosomal dominant

CLCN1

AD

1

1

1

2

1

1

Myotonic dystrophy 1 (DM1)

DMPK

AD

94

147

107

188

55

46

Myotonic dystrophy 2 (DM2)

CNBP

AD

1

2

2

4

2

2

Nail–patella syndrome (NPS)

LMX1B

AD

3

4

3

4

1

1

Nemaline myopathy 2 (NEM2)

NEB

AR

6

6

6

10

3

3

Nephrotic syndrome, type 1 (NPHS1)

NPHS1

AR

1

3

3

7

1

0

Nephrotic syndrome, type 2 (NPHS2)

NPHS2

AR

1

1

1

1

1

1

Nephrotic syndrome, type 5

LAMB2

AR

1

2

2

4

2

1

Neurofibromatosis, type I (NF1)

NF1

AD

51

90

80

123

46

41

Neurofibromatosis, type II (NF2)

NF2

AD

7

10

9

17

7

7

Neuropathy, hereditary sensory and autonomic, type III (HSAN3)

IKBKAP

AR

13

19

17

28

9

9

Neuropathy, hereditary sensory and autonomic, type VI (HSAN6)

DST

AD

1

2

2

2

2

2

Neutropenia, severe congenital, 1, autosomal dominant (SCN1)

ELANE

AD

1

1

1

1

1

1

Niemann–Pick disease, type A

SMPD1

AR

3

5

3

6

2

2

Nijmegen breakage syndrome (NBS)

NBN

AR

1

1

2

2

1

1

Noonan syndrome 1 (NS1)

PTPN11

AD

5

7

7

9

4

3

Norrie disease (ND)

NDP

XL

5

8

6

12

2

Omenn syndrome

RAG1

AD

2

6

5

12

1

1

Optic atrophy 1 (OPA1)

OPA1

AD

3

5

5

9

1

1

Ornithine transcarbamylase deficiency

OTC

XL

11

24

19

32

11

10

2

Osteogenesis imperfecta, type I (OI1)

COL1A1

AD

24

61

44

72

Osteogenesis imperfecta, type II (OI2)

COL1A2

AD

5

5

5

5

3

2

Osteogenesis imperfecta, type IX (OI9)

PPIB

AR

1

2

2

4

2

2

Osteopathia striata with cranial sclerosis (OSCS)

AMER1

XL

1

1

1

1

17

1

17

1

Osteopetrosis, autosomal recessive 1 (OPTB1)

TCIRG1

AR

5

7

7

13

3

3

Pachyonychia congenita 3 (PC3)

KRT6A

AD

1

2

2

2

2

1

PRSS1

AD

1

2

1

1

Paraganglioma and gastric stromal sarcoma

SDHB

AD

1

1

0

0

0

0

Paramyotonia congenita of von Eulenburg (PMC)

Pancreatitis, hereditary (PCTT)

SCN4A

AD

3

3

3

4

3

2

Pelizaeus–Merzbacher disease (PMD)

PLP1

XL

1

7

1

12

10

15

7

7

Periventricular nodular heterotopia 1 (PVNH1)

FLNA

XL

1

3

3

5

2

1

Peroxisome biogenesis disorder 1A (Zellweger) (PBD1A)

PEX1

AR

3

3

3

6

3

3

Peroxisome biogenesis disorder 2A (Zellweger) (PBD2A)

PEX5

AR

1

2

2

4

0

0

Peroxisome biogenesis disorder 3A (Zellweger) (PBD3A)

PEX12

AR

1

3

3

4

2

1

Peroxisome biogenesis disorder 5A (Zellweger) (PBD5A)

PEX2

AR

1

4

3

5

2

2

STK11

AD

4

9

7

9

6

4

Pfeiffer syndrome

FGFR1

AD

2

2

2

4

2

2

Phenylketonuria (PKU)

Peutz–Jeghers syndrome (PJS)

PAH

AR

15

20

14

16

8

7

Platelet disorder, familial, with associated myeloid malignancy (FPDMM)

RUNX1

AD

1

1

1

1

1

1

Pleuropulmonary blastoma (PPB)

DICER1

AD

1

1

1

1

1

1

Polycystic kidney disease 1 (PKD1)

PKD1

AD

48

84

64

98

37

34

Polycystic kidney disease 2 (PKD2)

PKD2

AD

7

10

9

15

3

3

Polycystic kidney disease, autosomal recessive (ARPKD)

PKHD1

AR

16

29

26

42

17

16

Polymicrogyria, bilateral frontoparietal (BFPP)

ADGRG1

AR

2

2

1

2

1

1

Polymicrogyria, bilateral frontoparietal (BFPP)

GPR56

AR

1

1

1

2

0

0

EXOSC3

AR

2

2

1

1

Popliteal pterygium syndrome (PPS)

IRF6

AD

2

2

1

2

1

1

Porphyria, congenital erythropoietic

Pontocerebellar hypoplasia, type 1B (PCH1B)

UROS

AR

1

1

1

1

1

1

PCCA,

Propionic acidemia

1

1

AR

3

3

3

5

2

2

F2 F5

AR

2

3

3

3

2

2

Pseudovaginal perineoscrotal hypospadias (PPSH)

SRD5A2

AR

1

2

2

4

1

1

Rap guanine nucleotide exchange factor 6 (RAPGEF6)

RAPGEF6

AD

1

2

3

4

3

1

1

1

2

2

1

1

PCCB Prothrombin deficiency, congenital; Factor V deficiency

Renal cell carcinoma, papillary, 1 (RCCP1)

MET

AD

Renal tubular acidosis, distal, autosomal recessive (RTADR)

ATP6V0A4

AR

1

1

2

3

2

1

Renal tubular dysgenesis (RTD)

ACE

AR

1

4

3

4

2

2 (Continued)

Table 2.1 (Continued) No.

No.

Type of

No.

No.

embryo

embryos

Pregnancy

No.

Conditions

Gene

inheritance

patients

cycles

transfers

transferred

%

deliveries

Restrictive dermopathy, lethal

ZMPSTE24

AR

2

2

2

3

1

1

Retinal dystrophy, early-onset, with or without pituitary dysfunction,

OTX2

AD

1

1

0

0

0

0

included XL

1

1

1

2

1

Retinitis pigmentosa 3 (RP3)

RPGR

XL

5

6

6

8

4

3

Retinitis pigmentosa 4 (RP4)

Retinitis pigmentosa 2 (RP2)

RHO

RP2

AD

3

5

2

4

1

0

1

Retinoblastoma (RB1)

RB1

AD

17

31

26

43

14

13

Retinoschisis 1, X-linked, juvenile (RS1)

RS1

XL

1

2

1

2

1

0

Rett syndrome (RTT)

MECP2

XL

3

5

4

4

3

1

Rhabdoid tumor predisposition syndrome 1 (RTPS1)

SMARCB1

AD

1

1

1

1

0

0

Rhesus blood group, D antigen (RHD)

RHD

AD

7

9

9

16

6

6

Sandhoff disease

HEXB

AR

4

6

5

8

4

4

Seckel syndrome 1 (SCKL1)

ATR

AR

1

1

2

2

0

0

Severe combined immunodeficiency, autosomal recessive

IL7R

AR

1

1

2

4

1

Severe combined immunodeficiency, autosomal recessive

RAG2

AR

2

5

4

5

3

3

Severe combined immunodeficiency, X-linked (SCIDX1)

IL2RG

XL

3

4

3

3

2

2

Short stature, idiopathic, X-linked (ISS)

SHOX

XL

2

2

2

3

2

1

2

Short-rib thoracic dysplasia 3 with or without polydactyly (SRTD3)

DYNC2H1

AR

1

1

1

1

1

1

Smith–Lemli–Opitz syndrome (SLOS)

DHCR7

AR

18

30

23

32

15

15

Sonic hedgehog (SHH)

SHH

AD

1

2

2

3

1

1

Sotos syndrome 1 (SOTOS1)

NSD1

AD

2

3

2

2

2

2

Spastic paraplegia 3, autosomal dominant (SPG3A)

ATL1

AD

1

1

1

1

1

1

Spastic paraplegia 4, autosomal dominant (SPG4)

SPAST

AD

6

10

8

12

7

5

Spherocytosis, type 2 (SPH2)

SPTB

AD

1

1

2

2

2

1

Spinal and bulbar muscular atrophy, X-linked 1 (SMAX1)

AR

XL

3

5

5

6

2

1

Spinal muscular atrophy, distal, autosomal recessive, 1 (DSMA1)

IGHMBP2

AR

2

3

2

4

1

1

Spinal muscular atrophy, type I (SMA1)

SMN1

AR

102

151

125

199

78

69

Spinocerebellar ataxia 1 (SCA1)

ATXN1

AD

4

7

8

4

Spinocerebellar ataxia 2 (SCA2)

ATXN2

AD

7

14

14

27

6

8

Spinocerebellar ataxia 6 (SCA6)

CACNA1A

AD

2

5

2

6

3

1

1

4

Spinocerebellar ataxia 7 (SCA7)

ATXN7

AD

2

3

3

7

2

1

Spinocerebellar ataxia 8 (SCA8)

ATXN80S

AD

1

1

1

1

1

1

Spondyloepiphyseal dysplasia tarda, X-linked (SEDT)

TRAPPC2

AD

1

1

2

2

1

1

Stargardt disease 1 (STGD1)

ABCA4

AR

4

10

5

6

2

2

Stickler syndrome, type I (STL1)

Col2A1

AD

4

4

3

5

2

2

Stickler syndrome, type II (STL2)

COL11A1

AD

2

7

6

15

1

1

Stickler syndrome, type II (STL2)

COL18A1

AR

1

1

1

1

1

1

Succinic semialdehyde dehydrogenase deficiency (SSADHD)

ALDH5A1

AR

3

4

4

9

2

2

Sulfocysteinuria

SUOX

2

1

1

AR

1

1

2

Supranuclear palsy, progressive, 1 (PSNP1)

MAPT

AD

2

3

3

5

1

Surfactant metabolism dysfunction, pulmonary, 3 (SMDP3)

ABCA3

AR

1

2

2

4

1

1

Symphalangism, proximal (SYM1)

NOG

AD

1

3

3

7

2

2

Tay–Sachs disease (TSD)

HEXA

AR

25

46

29

52

19

1

17

Telangiectasia, hereditary hemorrhagic, of Rendu, Osler, and Weber (HHT)

ENG

AD

4

11

6

7

3

3

Telangiectasia, hereditary hemorrhagic, type 2 (HHT2)

ACVRL1

AD

4

8

7

8

4

4

Temtamy syndrome (TEMTYS)

C12orf57

AR

1

1

1

2

0

0

Thrombocytopenia-absent radius syndrome (TAR)

RBM8A

AR

4

6

5

7

4

Treacher Collins syndrome 1 (TCS1)

TCOF1

AD

6

8

8

14

7

7

Treacher Collins syndrome 2 (TCS2)

POLR1D

AD

1

1

1

1

0

0

Tuberous sclerosis 1 (TSC1)

TSC1

AD

20

30

27

Tuberous sclerosis 2 (TSC2)

TSC2

AD

8

14

10

14

5

4

Tyrosinemia, type I (TYRSN1)

FAH

AR

1

7

7

13

5

3

Ulnar–Mammary syndrome (UMS)

TBX3

AD

1

3

3

4

1

1

52

16

4

14

Usher syndrome, type I (USH1)

MYO7A

AD

1

3

2

2

1

1

Usher syndrome, type IF (USH1F)

PCDH15

AR

2

4

4

6

4

2

USH2A

AR

2

2

Usher syndrome, type IIC (USH2C)

ADGRV1

AR

1

1

1

2

1

1

Usher syndrome, type IIC (USH2C)

Usher syndrome, type IIA (USH2A)

GPR98

AR

1

3

1

4

0

5

0

6

0

0

3

3

3

Van der Woude syndrome 1 (VWS1)

IRF6

AD

3

3

3

Von Hippel–Lindau syndrome (VHL)

VHL

AD

19

25

21

30

15

Waardenburg syndrome, type 2A (WS2A)

MITF

AD

2

6

6

6

4

4

Wilson disease

ATP7B

AR

3

3

3

5

3

2

Wiskott–Aldrich syndrome (WAS)

WAS

XL

6

15

13

20

9

8

Wolfram syndrome 1 (WFS1)

WFS1

AR

1

2

1

1

1

1

Xeroderma pigmentosum, complementation group g (XPG)

ERCC5

AR

1

1

0

0

0

0

3463

5869

4683

7443

2644

2332

1.59

56.4%

TOTAL

14

118 Genetic Disorders and the Fetus

MI

MII

N

N N

NN CF

CF CF

N CF CF

CF Affected Crossover

MII

CF CF

MII CF

N

N

CFN

CFN N

N

CF

C-1

Affected C-2 CFN CF CF N

CF N

N

Unaffected

Unaffected

developmental potential of the resulting embryo, 343 biopsied and 445 nonbiopsied mouse embryos were compared for the percentage of embryos reaching the blastocyst stage.36 The results of PGT-M performed by polar body biopsy, representing the world’s largest series, is shown in Table 2.2. A total of 1,016 PGT-M cycles were performed, for 538 autosomal recessive, 191 autosomal dominant, and 287 X-linked disorders. Of 1,016 cycles initiated, 838 (82.5 percent)

Figure 2.1 Scheme demonstrating the principle of preimplantation genetic analysis by sequential DNA analysis of the first and second polar body, using the cystic fibrosis (CF) locus as an example. Source: Verlinsky Y, Kuliev AMA. Preimplantation genetic diagnosis. In: Milunsky A, Milunsky JM, eds. Genetic disorders and the fetus: diagnosis, prevention and treatment, 6th edn. Oxford, UK: John Wiley & Sons, 2010.

resulted in transfer of 1,656 embryos (1.98 embryos per transfer on the average), 349 (41.6 percent) clinical pregnancies, and 385 babies born. Only two misdiagnoses were observed in the case of PGT for fragile-X syndrome and muscular dystrophy, which were due to consented transfer of additional embryo with insufficient marker analysis to exclude the probability of allele dropout (ADO) (see later). The example of PGT-M by polar body sampling is shown in Figure 2.2.

Table 2.2 Clinical outcome of PGT-M performed by polar body approach. Conditions/mode of inheritance/ sampling type

Patient

Cycles

Embryo

No.

transfer

embryos

Spontaneous Pregnancy

abortions

Baby

Autosomal recessive Polar bodies

76

131

99

204

36

10

36

Polar bodies + blastomere/blastocyst

254

407

344

701

143

21

168

Subtotal

330

538

443

905

179

31

204

21

Autosomal dominant Polar bodies

29

52

40

84

22

4

Polar bodies + blastomere/blastocyst

79

139

122

233

49

7

61

108

191

162

317

71

11

82

20

Subtotal X-linked Polar bodies

39

86

63

110

22

4

Polar bodies + blastomere/blastocyst

116

201

170

324

77

12

79

Subtotal

155

287

233

434

99

16

99

Total

593

1016

838

1656

349 (41.6%)

58 (17%)

385

Preimplantation Genetic Testing 119

CHAPTER 2

I. 160 158 166 176 111 114 DEL N DOB 11/1984 DEL N DEL N DEL N 108 110 PGT 144 158

II.

III.

PGT

D16S521 D16S525 D16S3395 TSC2 - DELETION D16S664 D16S3082

1 2 TSC2 FET 1 FET 2 Sequential polar body analysis 1

3

5

6

7

8

9 10 11 12 13 14 15 16 17 18 10 AFFECTED 5 NORMAL

Oocyte genotype Trophectoderm analysis Trophectoderm karyotype

160 166 111 DEL DEL DEL DEL 108 144 M

160 166 111 DEL DEL DEL DEL 108 144 M

158 176 114 N N N N 110 158 N

5

160 166 111 DEL DEL DEL DEL 10S 144 M

158 176 114 N N N N 110 158 N

160 166 111 DEL DEL DEL DEL 10S 144 M

160 166 111 DEL DEL DEL DEL 10S 144 M

160 166 111 DEL DEL DEL DEL 10S 144 M

160 166 111 DEL DEL DEL DEL 10S 144 M

7

158 158 176 176 114 114 N N N N N N N N 110 110 158 158 N N

12

FA

160 166 111 DEL DEL DEL DEL 10S 144 M

158 176 114 N N N N 110 158 N

160 166 111 DEL DEL DEL DEL 110 144 MR

160 166 111 DEL DEL DEL DEL 10S 144 M

Embryo #7 46, XY FET-1

17 Embryo #12 46, XX FET-2

45, –14, XX 46,XY FET 1

46,XX FET 2

Figure 2.2 Preimplantation genetic testing for de novo tuberous sclerosis* type II deletion (TSC2 gene, exon 7–10 deletion 16p13.3) and preimplantation genetic testing for aneuploidies by next-generation sequencing (NGS). Of 15 oocytes tested by polar body analysis, ten were affected and five free of deletion. The embryos deriving from deletion-free oocytes were tested for aneuploidy by NGS; three of these were euploid (embryos 7, 12, and 17) and one (embryo 5) with monosomy 14. Two of the mutation-free euploid embryos (embryos 7 and 12, the NGS

Preimplantation genetic testing based on embryo biopsy

Despite clear advantages, the polar body approach does not provide diagnosis of the paternal alleles and the gender of the embryo and therefore cannot be used to avoid the transfer of male embryos in cases of the X-linked disorders, unless specific diagnosis can be achieved on oocytes using the polar body approach (see later). The fact that the genotype of the oocyte is inferred from the genotype of the polar body, rather than determined directly is another weakness. In these situations, embryo biopsy becomes the much more comprehensive approach, with blastocyst biopsy currently being a standard procedure. The first clinical application of embryo biopsy for PGT by Handyside et al.19 was performed at the cleavage stage for X-linked disorders by gender determination.19 The study of the viability

46,XX

results of which are shown bottom right) were transferred in a frozen cycles, resulting in a twin pregnancy and birth of two unaffected children free from deletion. *Tuberous sclerosis complex is an autosomal dominant multisystem disorder characterized by hamartomas in multiple organ systems, including the brain, skin, heart, kidneys, and lung. Central nervous system manifestations include epilepsy, learning difficulties, behavioral problems, and autism. The affected mother had had epilepsy since 3 months old and lympho-angio-leiomyomatosis.

of the biopsied pre-embryos did not reveal any detrimental effect of these procedures: it was shown that more than 70 percent of the manipulated embryos reached blastocyst stage, with no significant reductions in cell number and energy substance (glucose and pyruvate) uptake.37 Embryo biopsy, initially applied at the cleavage stage, has become a method of choice in most centers, resulting in the birth of thousands of children free of genetic disorders.21–25 Nevertheless, some programs have demonstrated significant detrimental effect of the procedure, especially when instead of one blastomere two were removed, or even a single blastomere removed in inexperienced hands (see later). Also, there were problems due to the high rate of allele dropout and the high frequency of mosaicism at this stage (see later). The switch to blastocyst biopsy largely solved these problems and became standard. In

120 Genetic Disorders and the Fetus

fact, blastocyst biopsy was first introduced in the 1960s in rabbits by Gardner and Edwards.38 As the number of cells in human blastocyst increases up to more than 100, a few cells (approximately five cells) are removed from IVF embryos without affecting viability. Another advantage of this approach is that trophectoderm cells are biopsied without affecting the inner cell mass, from which the embryo is derived. Initially the viability of biopsied blastocysts in vitro was studied using morphologic criteria and the patterns of human chorionic gonadotropin (hCG) secretion. Hatching was observed in 38.5 percent of blastocysts, with hCG detected first on day 8, peaking at day 10, and still detectable in some blastocysts at day 14.39 For the individual blastocysts, the pattern of hCG secretion correlated with the assessment of morphology.40 Blastocyst biopsy for clinical purposes was first attempted over 20 years ago41–43 and is currently a PGT standard.44, 45 This is also the method used in uterine lavage, which may soon appear as a realistic approach for PGT without IVF.46 Both mechanical44–46 and laser techniques were used for blastocyst biopsy, which has become a method of choice in most centers, also resulting in improved pregnancy rates, particularly in frozen PGT cycles. The advantage of blastocyst biopsy over cleavage-stage sampling was demonstrated by a well-designed randomized controlled study.47 The procedure is performed as follows: On day 5 of embryo development, when the blastocyst begins to herniate through the zona pellucida, several herniated trophectoderm cells are removed by smooth aspiration into a biopsy pipette with an internal diameter of 30 μm through the zona pellucida, which is opposite the inner cell mass. To break down the tight junctions between trophectoderm cells, three laser shots are applied (with a duration of 0.7 ms pulse for each shot). In the selection of embryos for blastocyst biopsy, poor-quality blastocysts and those with early stage herniation are avoided. It is also possible to perform blastocyst biopsy without the application of laser in order to avoid potential damage to the cells. The technique depends on the use of the force of surface tension on the boundary between the biopsy drop and the mineral oil culture medium, instead of laser pulses. The method can be applied only on grades

2 to 2–3 of the day 5 blastocysts, because the tight junction between trophectoderm cells of the early blastocyst (grades 2 and 2–3) is not as strong as the cell connection in the more advanced blastocyst (grades 3–4, 4, and higher). Grade 2 and 2–3 blastocysts are placed into 5 μL equilibrated culture media drops covered by 2 mL of equilibrated mineral oil, held by holding the pipette on the left side, while the biopsy pipette (interior diameter 25 μm) on the right side is used to suck 5–10 extruded trophectoderm cells into the pipette. The embryo is then moved onto the right edge of the biopsy drop with the biopsy pipette, moving slowly toward the inside of the oil environment, making a cytoplasm bridge between blastocyst and sample thinner, until it is finally removed and placed into a separate 5 μL culture media drop in the same dish. This procedure results in nonsticky trophectoderm samples with no evidence of damaged nuclei, with improved amplification efficiency.48 As the time for analysis is limited by the implantation window, which is less than 24 hours, the technique of vitrification of biopsied blastocysts is now routine, allowing sufficient time before transfer of the tested embryos in a subsequent freeze–thaw cycle. It appears that this approach has also improved implantation and pregnancy rates, which could also be explained by the better receptivity of the uterus in unstimulated cycles. The other advantage of the method is that the embryo has already passed the natural self-correction mechanisms, overcoming the natural errors of the cleavage stage, thus enabling the diagnosis of only persisting abnormalities. Prospects for noninvasive preimplantation genetic testing

Although there is no evidence of the detrimental effect of biopsy procedures on embryo viability, the potential for damage cannot be excluded, so the development of noninvasive preimplantation genetic testing (NIPGT) is of increasing importance. The fact that DNA fragments of approximately 150 bp are present in cell-free DNA of blastocoele fluid and spent culture medium, is the basis of the concept that NIPGT can be developed analogous to noninvasive prenatal testing (NIPT), because these DNA may originate

CHAPTER 2

from breakdown of nuclear DNA derived from cells damaged at biopsy or undergoing apoptosis during cell division. Available reports suggest that the technique may in future be applicable for preselection of euploid embryos for transfer.49–58 Although the usefulness of blastocoele fluid for PGT by different groups lacks consensus, its use has been proposed to identify at-risk embryos from younger patients who otherwise have no accessible indication for PGT-A.49, 50 The other approach, based on the use of cell-free DNA in culture media, represents the genuine noninvasive approach analogous to monitoring cell-free DNA in maternal plasma during pregnancy,51–58 with the concordance studies showing progress. In one of these studies, the test was offered to infertility patients presenting for PGT-A in the format of a clinical trial. Enough DNA for testing was detected in 88% of cases, with 80% concordance to biopsy results.55 In another prospective study, blastocoele samples and spent culture medium samples were compared to diagnostic biopsy samples that were processed for PGT-M and PGT-A. Overall results demonstrated that neither blastocoele samples nor spent medium were sufficiently robust approaches for aneuploidy or single-gene disorders and cannot be applied clinically until the risk of maternal contamination can be excluded. This risk is particularly high in spent culture medium samples due to maternal cumulus DNA contamination.56 In the other study DNA in spent medium was shown to be detectable on day 3, but more reproducibly on day 5, with concordances of 65 and 70 percent with biopsy samples, which are not high enough for practical application.57 Although the origin of the DNA in spent culture medium is not clear, it was postulated that results of the test may have a prognostic utility and better prediction of reproductive outcome if euploid embryos with euploid spent media results are transferred. This is in contrast to the euploid embryo transfer outcome with spent media showing imbalanced results.58 This is also in agreement with the blastocoele data, which suggested a significantly improved embryo transfer outcome for those euploid embryos whose blastocoele fluid has a higher quantity of DNA based on WGA.50 Thus, clearly more research is needed to validate the usefulness of spent culture medium and blastocoele fluid for PGT.

Preimplantation Genetic Testing 121

Preimplantation genetic analysis Initially, PGT was justified only for high-risk pregnancies. Maternal age was not expected to be an indication for such early testing, initially being considered even a contraindication to PGT. However, the development and improvement of the methods for sampling and genetic analysis have made use of PGT for chromosomal disorders a reality. Despite continuing discussion of the impact of PGT-A, it is used routinely worldwide as a tool in assisted reproduction technologies, especially for improving the effectiveness of IVF in poor-prognosis patients, particularly in carriers of chromosomal rearrangements. As a result, the majority of PGT cycles are still done for PGT-A.21–25 Single-gene disorders

DNA analysis for preconception and preimplantation diagnosis is well established, which enables genetic analysis of minute quantities of DNA obtained from a single or few cells.12, 19, 20 Because this also increases the chance of DNA contamination in PGT, decontamination procedures were applied in the initial stages, based on elimination of double-stranded DNA sequences,59 excluding also possible contamination with the embryology and PCR reagents, such as water, salt solutions, oligonucleotides, and Taq polymerase. The major source of contamination in PGT is still cellular contamination, such as cumulus cells, spermatozoa, or cell fragments, which might be amplified simultaneously with polar bodies or embryo biopsies, creating the possibility for erroneous testing. Because potential misdiagnosis of PGT may be caused by sperm DNA contamination, it is currently a routine IVF practice to perform PGT-M for single-gene defects following microsurgical fertilization by intracytoplasmic sperm injection (ICSI). Nevertheless, the major source of misdiagnosis is preferential amplification or allele-specific amplification, referred to as allele dropout (ADO), which may happen in single-cell genetic analysis. As much as 8 percent of ADO was observed in PCR analysis of the first polar bodies, reaching over 20 percent in blastomeres.60 False-negative diagnoses have been observed in PGT for X-linked disorders, myotonic dystrophy, and cystic fibrosis

122 Genetic Disorders and the Fetus

(CF), at the initial stage of PGT clinical application.3, 22, 23, 25, 35, 59 Clearly, the failure to detect one of the mutant alleles in compound heterozygous samples due to ADO will lead to misdiagnosis. However, this is no longer a problem with the application of protocols for simultaneous detection of the causative gene together with highly polymorphic markers closely linked to the gene tested.59 With simultaneous testing of a sufficient number of linked markers amplified together with the gene in question, the risk of misdiagnosis may be substantially reduced or even practically eliminated. The protocol involves a multiplex nested PCR analysis, with the initial first-round PCR reaction containing all the pairs of outside primers, followed by amplification of separate aliquots of the resulting PCR product with the inside primers specific for each site. Following the nested amplification, PCR products are analyzed by restriction digestion, real-time PCR, direct fragment size analysis, or mini-sequencing. Depending on the mutation being studied, different primer systems are designed with special emphasis on eliminating false priming to possible pseudogenes, for which purpose the first-round primers are designed to anneal to the regions of nonidentity with a pseudogene.25, 59 With the introduction of next-generation technologies and the use of WGA prior to DNA analysis, the risk of ADO is further increasing, presenting even more problems in achieving accurate diagnosis.61 To improve the reliability of the test, the use of multiple linked markers became even more important, with importance of not only excluding the presence of the mutant gene, but also confirming the presence of the normal allele(s). Although a sufficient number of informative closely linked markers are usually available for multiplex PCR, this might not be the case in performing PGT by conventional PCR analysis in some ethnic groups.59 Currently available protocols allow an accurate PGT for complex cases, requiring testing for two, three, and even more different mutations. PGT generally requires knowledge of sequence information for Mendelian diseases, but may also be performed when the exact mutation is unknown. With the expanded use of single nucleotide polymorphisms (SNPs), linkage analysis allows PGT for any monogenic disease, irrespective of the

availability of specific sequence information.59–64 This is a more universal approach to track the inheritance of the mutation without actual testing for the gene itself, such as in karyomapping.65 On the other hand, a specific diagnosis is required for X-linked disorders, which may be performed by polar body analysis to preselect the embryos deriving from mutation-free oocytes which may be transferred irrespective of gender or the paternal genetic contribution.66 Polar body analysis (see Table 2.2 and Figure 2.2) also provides the prospect of pre-embryonic diagnosis, which is required in many population groups where objection to the embryo biopsy procedures makes PGT nonapplicable. We performed the first pre-embryonic genetic diagnosis for Sandhoff disease in a couple with a religious objection to embryo destruction.67 Although pre-embryonic genetic diagnosis was previously attempted by first polar body testing,68–71 it is not actually sufficient for accurate genotype prediction without second polar body analysis, as shown in Figure 2.1. It is understood that for pre-embryonic testing the second polar body analysis should be done prior to pronuclei fusion (syngamy), to ensure that only zygotes originating from mutation-free oocytes are allowed to progress to embryo development and to be transferred, avoiding the formation and possible discard of any unaffected embryo. A particular challenge is also presented by PGT for mitochondrial diseases, which still cannot be done reliably. A novel approach has been made to transfer a nuclear genome from the pronuclear stage zygote of an affected woman to an enucleated donor zygote, or to transfer the metaphase II spindle from an unfertilized oocyte of an affected woman to an enucleated donor oocyte.72 As seen from Table 2.1, PGT is no longer restricted to conditions presented at birth; it is gradually expanding to include common diseases with genetic predisposition, such as cancers, performed in 10.5 percent of PGT-M cycles, or nongenetic indications (7.2 percent of cases), such as PGT-HLA with the purpose of stem cell therapy of affected siblings in the family.62 Here we discuss the application of PGT-M to a wider range of disorders, including conditions determined by de novo mutations (DNMs), genetic predisposition for late-onset disorders, and preimplantation HLA matching (Table 2.1).

CHAPTER 2

De novo mutations

PGT is presently applicable to couples who, although they may themselves be noncarriers of a mutation, have been found to have a DNM in their gonads although there is no family history of the genetic disease, or the disease is first diagnosed in one of the parents or their affected children (see Figure 2.2). As neither the origin nor relevant haplotypes may be available for tracing the inheritance of such mutations in single cells biopsied from embryos or in oocytes, the main emphasis is on the identification of the mutation and/or relevant haplotypes enabling mutation detection. Accordingly, PGT strategies for DNM depend on their origin. DNA analysis of the parents and affected children prior to PGT is required for verification of the mutation and polymorphic markers through single sperm testing and polar body analysis, thereby providing the normal and mutant haplotypes to trace the mutation. If the origin of the mutation is paternal, confirmation is first sought on the paternal DNA from blood and total sperm, and then by single sperm typing to determine the proportion of sperm with DNM and relevant normal and mutant haplotypes. It is also useful to test the relevant linked markers for the partner, to exclude misdiagnosis due to possible shared maternal and paternal markers. Where the origin of the mutation is maternal, polar body testing is the method of choice, providing the normal and mutant maternal haplotypes. Again to exclude misdiagnosis caused by possible shared paternal and maternal markers, the relevant paternal haplotypes are established through a single sperm typing. If the mutation was first detected in children, both the maternal and paternal haplotypes are established as described. The other important phenomenon detected in PGT for DNM is gonadal mosaicism, which can be detected in either parent. Although the strategies may differ depending on the type of DNM inheritance, the general approach involves the identification of DNM origin and search for a possible gonadal mosaicism and relevant parental haplotypes. Despite the complexity of PGT for DNM, these strategies may be applied in clinical practice with extremely high accuracy without the traditional requirement for family data, which is not always available. Since the report of our first systematic experience of PGT for 152 families with different

Preimplantation Genetic Testing 123

genetic disorders,73 we have performed 526 cycles from 283 couples for 81 different de novo conditions, resulting in 270 clinical pregnancies and the birth of 234 unaffected children, with no misdiagnosis.48 Late-onset disorders

PGT for late-onset disorders with genetic predisposition was first applied for a couple with inherited cancer predisposition, determined by p53 tumor suppressor gene mutations,74 which are known to determine a strong predisposition to many cancers. Traditionally, these conditions have not been considered as an indication for prenatal diagnosis that would lead to pregnancy termination, which is not justified on the basis of genetic predisposition. Rather, the possibility of choosing embryos free of genetic predisposition for transfer would obviate the need for considering pregnancy termination, as only potentially normal pregnancies are established. Although the application of PGT for these conditions is still controversial, it has been performed for an increasing number of disorders with genetic predisposition that present beyond early childhood and may not even occur in all cases, including inherited cancers and heart disease.6, 7, 25, 74–77 We have performed a total of 874 cycles for 56 different forms of cancers, the most frequent being breast cancer (284 cycles) caused by BRCA1 (159 cycles) and BRCA2 (125 cycles) mutations. A total of 199 PGT cycles for BRCA1/2 resulted in transfer of one or two embryos, yielding 131 pregnancies and birth of 134 children free from genes predisposing to breast cancer.48 The other largest group of cancers for which PGT was performed was neurofibromatosis type 1 and type 2 (NF1/2), for which 103 cycles resulted in transfer of 138 genetic predisposition-free embryos in 88 cycles, 53 clinical pregnancies, and 55 children born free from genes predisposing to neurofibromatosis. Other cancers representing frequent indications for PGT were different types of Fanconi anemia (83 cycles), colorectal cancer (52 cycles), tuberous sclerosis types 1 and 2 (44 cycles) (see Figure 2.2), familial adenomatous polyposis (42 cycles), multiple endocrine neoplasia (34 cycles), and retinoblastoma (31 cycles); under 30 PGT cycles were performed for various other cancers.

124 Genetic Disorders and the Fetus

Overall, 966 genetic predisposition-free embryos were transferred in 634 cycles, resulting in 387 (61.0%) clinical pregnancies and birth of 407 children free from the risk of developing these cancers.25, 48 The other emerging PGT-M indication has been inherited cardiac disease, for which 109 cycles were performed relating to 23 different diseases. The most frequent indications were familial hypertrophic cardiomyopathy, CMH4 (22 cycles), dilated cardiomyopathy, CMD1A (17 cycles), Holt–Oram syndrome, HOS (8 cycles), acyl-CoA dehydrogenase very-long-chain deficiency, ACADVLD (6 cycles), familial hypertrophic cardiomyopathy 1, CMH1 (6 cycles), long QT syndrome 1, LQT1 (6 cycles) and Noonan syndrome 1, NS1 (6 cycles); PGT for another 16 cardiac conditions were performed in five or less number of cycles. Overall, 123 embryos free of genes predisposing to cardiac disease were transferred in 89 cycles (1.38 embryos per transfer on the average), resulting in 55 clinical pregnancies (61.7 percent) and birth of 54 children free from inherited predisposition to these cardiac diseases. If not prevented, many of these conditions may manifest despite presymptomatic diagnosis and follow-up, with their first and only clinical occurrence being a premature or sudden death.78 The couples at risk for producing progeny with inherited cardiac disease usually request PGT prospectively, with no previous pregnancies attempted, given one of the partners being a carrier of the specific mutation. Many couples already going through IVF for fertility treatment may have questions about the implications of genetic susceptibility factors for offspring, and the appropriateness of using PGT in testing for susceptibility to inherited cardiac disease.25, 48 Of special interest are PGT indications for late-onset disorders with inherited predisposition to neurological disorders, including neurodegenerative conditions. A total of 960 PGT cycles were performed for these conditions, including 610 for intellectual disability, 210 for Huntington disease, 42 for different movement disorders, such as torsion or myoclonic dystonia, nine for Alzheimer disease, and nine for Prion disease. As many as 1,110 unaffected or genetic predisposition-free embryos were transferred in 738 cycles (1.5 embryos per transfer on average), yielding 412 clinical pregnancies and birth of 406 infants

unaffected or free of the genes predisposing to the above conditions.48 Thus, PGT provides a nontraditional option for patients who may wish to avoid the transmission of a mutant gene predisposing to late-onset disorders in their future children. Because such diseases that present beyond early childhood and even later may not be expressed in 100 percent of cases, the application of PGT for this group of disorders is still controversial. However, for diseases with no current prospect for treatment, PGT may still be offered as the only relief for at-risk couples. HLA typing

Preimplantation HLA typing is an attractive PGT indication. The first case of preimplantation HLA typing was performed in combination with PGT for Fanconi anemia complementation group C (FA-C), which resulted in a successful hematopoietic reconstitution in the affected sibling by transplantation of stem cells obtained from the HLA-matched offspring resulting from PGT.5 To improve access to the HLA-identical bone marrow transplantation in sporadic bone marrow failures, this approach was then applied with the sole purpose of ensuring the birth of an HLA-identical offspring, not involving PGT, which also resulted in radical treatment of a sibling with a sporadic Diamond–Blackfan anemia (DBA) by stem cell transplantation from an HLA-identical child born following preimplantation HLA typing.79 Preimplantation HLA typing has become one of the most useful indications for PGT, performed currently with or without testing for the causative gene.79–88 Despite the ethical issues involved,80 preimplantation HLA typing procedures have so far been performed in hundreds of cases with affected children requiring HLA-compatible stem cell transplantation, including thalassemia, Fanconi anemia, Wiskott–Aldrich syndrome, X-linked adrenoleukodystrophy, X-linked hyper-IgM syndrome, X-linked hypohidrotic ectodermal dysplasia with immune deficiency, X-linked chronic granulomatous disease, cancer syndromes, incontinentia pigmenti, leukemias, and inherited and sporadic forms of DBA.81–91 We applied PGT-HLA in 485 cycles, including HLA typing alone, or combined with PGT-M for 35 different conditions. Overall, 424 HLA-matched unaffected embryos were detected and transferred

CHAPTER 2

in 291 cycles, resulting in 125 clinical pregnancies and birth of 117 HLA-matched children, as potential donors for their siblings.25, 48, 92 Although the majority of cases were performed for thalassemia, this approach has a great life-saving potential for affected siblings with congenital immunodeficiency. We performed 135 PGT cycles for 18 different immunodeficiency conditions, resulting in the birth of 54 children free of immunodeficiency, stem cell donors for transplantation treatment of affected siblings, with a total cure, such as Fanconi anemia and hyper-IgM syndrome.48 Similar experience has been reported from other large series, such as from Istanbul: 626 PGT-HLA cycles for 312 couples were performed (122 HLA only and 504 with PGT-M), resulting in the birth of 128 thalassemia-free children. Stem cells of 66 of these children were used for cord blood or bone marrow transplantation, which resulted in successful bone marrow reconstitution in all but two of them (transplantation treatment of the remaining 57 sibling pending).93, 94 Chromosomal disorders

The theoretical rate of chromosomally abnormal embryos at fertilization is approximately 40 percent, taking into account both the rate of aneuploidies in oocytes and sperm and fertilization-related abnormalities.95, 96 Mouse data show that most aneuploidies, although compatible with cleavage, are lost during implantation.97, 98 Additional losses of chromosomally abnormal embryos occur after implantation, clinically recognized as spontaneous abortions, more than half of which are caused by chromosomal abnormalities. As a result of this selection against chromosomal abnormalities before and after implantation, only 0.65 percent of newborns have chromosomal disorders, many of which lead to serious disability and early death (see also Chapter 1).

Prevalence and origin of chromosomal errors A wide range in the frequency of chromosomal aneuploidy in human oocytes has been reported (17–70 percent), but most of these studies have been performed on poor-quality oocytes left over after the failure of IVF attempts. A high rate of hypohaploidy observed in oocytes was considered to be artificially induced by spreading techniques,

Preimplantation Genetic Testing 125

so aneuploidy rate was calculated by doubling the number of hyperhaploid oocytes. This ignores chromatid malsegregation and/or chromosome lagging events, contradicting the results of the observation that the rate of hypohaploidy is higher than the rate of hyperhaploidy.99 Cytogenetic analysis of unfertilized oocytes was also improved by parthenogenetic activation of human oocytes.100 As mentioned, an attempt at noninvasive cytogenetic analysis of oocytes was undertaken in the early 1980s through visualization of the second polar body chromosomes by transplanting the polar body into a fertilized egg.33 The success rate of visualization of polar body chromosomes was then improved by different methods, demonstrating the practical implication of polar body analysis for chromosomal errors originating from maternal meiosis,101–103 in contrast to the report on the uselessness of the first polar body for this purpose.104 Various approaches were explored in the attempt to visualize the chromosomes of the first and second polar bodies, as well as of the biopsied blastomeres, including nuclear transfer, electrofusion, and chemical methods.102, 105, 106 However, the major progress in chromosome analysis of oocytes and embryos was achieved with introduction of the fluorescence in situ hybridization (FISH) technique,107–113 microarray technology, and NGS. Our meiosis data based on the analysis of 22,986 oocytes detected 9,812 aneuploid oocytes (46.8 percent), originating comparably from the first and second meiotic divisions. Overall, meiotic division errors were observed in 33.1 percent of oocytes in meiosis I, 38.1 percent in meiosis II, and 28.8 percent in both. Although the aneuploidy rate in embryos is comparable to that in oocytes, the types of anomalies in the oocytes and embryos were significantly different,114–116 also showing inconsistency between the expected and observed frequency of some types of aneuploidies. In our current practice of PGT-A, the analysis of 2,922 blastocysts, 56.0 percent of embryos were aneuploid, comprising 13.0 percent monosomy, 13.0 percent trisomy, 8.0 percent numerical mosaic, 14.0 percent segmental mosaic, and 8.0 percent complex errors.48 It is of interest that no age dependence was revealed for monosomies in embryos, suggesting that the rate of monosomies detected in embryo by PGT-A may be of artefactual nature.

126 Genetic Disorders and the Fetus

A possible explanation for this discordance is that the majority of monosomies detected in embryos are derived from mitotic errors, assuming technical causes are excluded. In fact, a significant proportion of the cleavage-stage monosomies appeared to be euploid after their reanalysis.117, 118 No monosomies, except monosomy 21, are detected after implantation, so either they are eliminated before implantation or have no biological significance, reflecting the poor viability of the monosomic embryos and their degenerative changes. In one relevant study the progression and survival of different types of chromosome abnormalities were followed up in 2,204 fertilized oocytes.119 A variety of chromosome abnormalities was detected, including many types of errors not recorded later in development. However, these appeared to be tolerated until activation of the embryonic genome, after which there were declines in frequency. Nevertheless, many aneuploid embryos still successfully reach the blastocyst stage, even if some chromosome errors present during preimplantation development are not seen in later pregnancy.

Aneuploidies As seen from the previous discussion, without the detection and avoidance of the transfer of chromosomally abnormal embryos, there is at least 50 percent chance of loss during implantation or postimplantation development. In addition to the clear benefit of avoiding the transfer of aneuploid embryos, which contributes to improvement of pregnancy outcome of poor-prognosis IVF patients, this should improve the overall standard of medical practice, upgrading the current selection of embryos by morphologic criteria into aneuploidy testing. This explains a widespread application of PGT-A aimed at the preselection of embryos with the highest developmental potential demonstrating a clinical benefit, in terms of the improved IVF outcome through improved implantation and pregnancy rates, reduction of spontaneous abortions and improved take-home baby rates in IVF patients of advanced reproductive age, repeated IVF failures, and recurrent spontaneous abortions.24, 25, 114–116, 120, 121 The failure to detect a positive effect of aneuploidy testing on reproductive outcome in a few studies may be due to possible methodologic deficiencies.122–124 Despite these methodological

shortcomings, which have been heavily criticized in the literature,125–127 the American Society for Reproductive Medicine Practice Committee did not favor transferring embryos without aneuploidy testing.128 This may mean the alternative of incidental transfer of chromosomally abnormal embryos, as every second oocyte or embryo obtained from poor-prognosis IVF patients is chromosomally abnormal and destined to be lost before or after implantation. In fact, only one in ten of chromosomally abnormal embryos may survive to recognized clinical pregnancy, 5 percent may survive to the second trimester, and only 0.5 percent reach birth. Thus, the majority of chromosomal abnormalities are eliminated before or during implantation, reflecting a poor implantation rate in poor-prognosis IVF patients, and explaining a high fetal loss rate in those patients without PGT-A. This has actually been demonstrated by testing products of conception from poor-prognosis non-PGT IVF patients, confirming the high prevalence of chromosomal aneuploidy in the absence of PGT-A. Of 273 cases tested, 64.8 percent had chromosomal abnormalities, up to 79 percent of which could have been detected and not transferred using PGT.129 Given these data, the current IVF practice of selecting embryos for transfer based on morphologic criteria may hardly be an acceptable procedure for poor-prognosis IVF patients. In addition to giving an extremely high risk of establishing an affected pregnancy from the onset, this will significantly compromise the very poor chances of these patients to become pregnant,130, 131 especially with the current tendency of limiting the number of transferred embryos to only a few or even one. Although culturing embryos to day 5 (blastocyst) before transfer may allow, to some extent, the preselection of developmentally more suitable embryos compared with day 3, some aneuploid embryos will still be capable of developing to the blastocyst stage.132, 133 These abnormal embryos will not be eliminated in the current shift to blastocyst transfer, and may implant and lead to fetal loss, compromising the outcome of pregnancies resulting from the implanted normal embryos in multiple pregnancies. In fact, multiple pregnancies represent a severe complication of IVF, which is currently avoided by preselection and transfer of a single aneuploidy-free blastocyst with

CHAPTER 2

the greatest developmental potential to result in a healthy pregnancy. However, contrary views also exist about safety, outcome, and efficacy.122–124, 134–137 Randomized controlled studies performed with introduction of next-generation technologies were able to quantify the clinical impact of preselection of aneuploidy-free zygotes, demonstrating the obvious benefit approximating a 15–20 percent increase in pregnancy rates, compared to embryos transferred solely based on morphological criteria, although this was not universal in all age groups. The first randomized controlled trial (RCT) using 24-chromosome analysis was performed in a series of 112 women randomized into two groups:138 transfer of a PGT-A embryo versus transfer of a morphologically normal embryo not biopsied or tested. Of 425 blastocysts tested, 45 percent (191/425) were with aneuploidy, resulting in a 71 percent pregnancy rate compared with 46 percent in the nonbiopsied control group of 389 blastocysts with normal morphology. In the other RCT, involving 72 cases, the transfer of euploid blastocysts resulted in 66 percent implantation and 85 percent delivery rates, compared to 48 percent and 68 percent, respectively, in the control group of 83 morphologically normal embryos but not tested for PGT-A.139 Another RCT did not find differences in pregnancy rates between single euploid embryo transfer and the transfer of two morphologically normal but untested embryos, but a 48 percent twin rate was observed in the latter compared to 0 percent in the single embryo transfer tested group.140 Significant differences between a PGT-A group and control groups were also demonstrated in an RCT performed using a cleavage-stage embryo biopsy.141 Thus, results of RCTs involving 24-chromosome platforms suggest that it is reasonable to inform assisted reproductive technology (ART) patients of advanced maternal age about the utility of PGT-A. The precise age range at which women would benefit is still under study, although the optimal outcome seems to be for the 35–39 age group, as suggested by trials conducted by the Society for Assisted Reproductive Technology (SART)142 and the STAR Study Group.143 The switch of aneuploidy testing from FISH to the next-generation technologies for 24-chromosome testing,144–163 allowing improved detection

Preimplantation Genetic Testing 127

of chromosomally abnormal oocytes and embryos, has, therefore, further confirmed the positive impact of avoiding aneuploid embryos from transfer. In addition to testing all 24 chromosomes, the switch from blastomere sampling to blastocyst biopsy has also contributed to the positive reproductive PGT-A outcome, as only established anomalies are tested. With progress in vitrification procedures, blastocyst biopsy coupled with transfer after freezing in a subsequent cycle has become the major approach for PGT, as it also involves a much higher uterine receptivity than in stimulated cycles. Blastocyst biopsy has also improved PGT accuracy, because instead of using single cells, a number of cells are used for analysis. Blastocyst biopsy and vitrification, coupled with 24-chromosome testing, also simplified the organizational aspects of PGT, because the samples can be processed without the time limits for genetic analysis, also allowing samples to be shipped to specialized centers for more sophisticated testing, if required. Present standards of PGT-A are presented in Figure 2.3. As seen from Figure 2.3, the gold standard for PGT-A is presently NGS, which is also the initial step in PGT-M, also using WGA.160–163 Compared with other PGT-A methods, NGS provides more accurate copy number variations for each chromosome and is, therefore, better able to identify the presence of mosaic aneuploidy within the blastocyst. The detection of mosaicism requires much higher resolution than that provided by array comparative genomic hybridization (array-CGH). This is achieved by NGS, which has improved accuracy of testing, especially in detecting copy number variations that contribute to the mosaicism detection rate. Different labs use different cut-off rates, but PGDIS recommendations currently recommend a 20 percent cut-off: embryos are considered nonmosaic euploid if nonmodel DNA proportions are below 20 percent. Nonmodel DNA over 80 percent is considered nonmosaic aneuploid. Between 20 and 80 percent are considered mosaic.164, 165 Figure 2.4 presents an example of mosaicism detected by NGS. The applicable commercially available kit for NGS is VeriSeqTM PGT Kit (Illumina). Karyomapping supplied by Illumina may also be used for PGT-A, but requires different equipment and reagents than NGS. The primary application of karyomapping is PGT-M.65 The other alternative NGS platform

128 Genetic Disorders and the Fetus

Next generation sequencing (NGS)

Chromosomal postion

8 9 10 11 12 13 14 15 16 17 18 2190 21 22 X Y

6 7

3

5

4

1

7 8 9 10 11 12 13 14 15 16 17 18 2190 21 22 X Y

6

5

4

3

1

Chromosomal postion

2

4.00 Mos45/46,XX, –2 Mosaic 3.60 3.20 2.80 Mosaic range (20–80%) 2.40 2.00 1.60 Mosaic range (20–80%) 1.20 0.80 0.40 PGT-A by NGS Can Detect mosaicism

45,XY,-16

2

6 7 8 9 10 11 12 13 14 15 16 17 18 2190 21 22 X Y

5

4

4.00 3.60 3.20 2.80 2.40 2.00 1.60 1.20 0.80 0.40 3

2

4.00 46,XX 3.60 3.20 2.80 2.40 2.00 1.60 1.20 0.80 0.40 1

Copy number

Blastocyst biopsy

Chromosomal postion

and cells still undergoing DNA replication, so the results per Figure 2.3 Present standards of preimplantation genetic analysis for aneuploidies (PGT-A). Twenty-four-chromosome embryo derive from proportion of normal (euploid) and aneuploidy testing by measurements of DNA content – not abnormal (aneuploid) DNA. number of cells. DNA content may include damaged cells

Copy number

Chromosomal position

Y

14 15 16 17 18 19 20 21 22 X

11 12 13

9

10

8

7

6

5

4

3

2

1

4.00 3.60 3.20 2.80 2.40 2.00 1.60 1.20 0.80 0.40

Figure 2.4 Mosaicism for monosomy 3 detected by next-generation sequencing (NGS). NGS results show a 50 percent mosaicism for monosomy 3, with all other chromosomes showing a normal pattern.

is Personal Genome Machine (PGM), developed by Thermo-Fisher Scientific. The commercially available kit for this platform is Ion ReproSeqTM PGS Kit (Thermo-Fisher Scientific). The major concern with NGS is that it is prone to ADO, because WGA must be performed as a first step to generate an adequate amount of DNA for analysis, which, as mentioned earlier, is still extremely inefficient in recovering all genomic sequences. So although NGS allows concomitant PGT-A and PGT-M, without simultaneous testing of a sufficient number of linked markers false-negative results cannot be excluded, which may then lead to misdiagnosis, especially in PGT

for dominant diseases. It can therefore be predicted that the technique should be performed with the use of SNP analysis for this purpose, or to work out the level of deep sequencing that can overcome the problem of ADO or develop more efficient WGA.61, 159

Structural rearrangements The impact of PGT is even higher in translocation patients, with considerable reduction in the spontaneous abortion rate after preimplantation testing, resulting in a corresponding increase in the take-home baby rate.130, 131 Although previous experience with PGT for chromosome structural

CHAPTER 2

Copy number

Preimplantation Genetic Testing 129

Chromosomal position

4.00 46, XX, t(6;18)(p21.3;p11.2)

3.60 3.20 2.80 2.40 2.00 1.60 1.20 0.80

X Y

15 16 17 18 1 209 21 22

11

12 13 14

9

10

8

7

6

5

4

3

2

1

0.40

Figure 2.5 Next-generation sequence-based testing for translocation 46,XX, t(6;18)(p21.3;p11.2) (derivative chromosome indicated by red arrows).

rearrangements (PGT-SR) was based on the use of the FISH technique, which is still applicable in some specific cases, the current standard is the utilization of array-CGH or NGS technologies, which improves accuracy of testing and also allows a combined PGT-A. An example of PGT-SR by NGS is shown in Figure 2.5. In our experience of 940 PGT-SR cycles, the comparison of reproductive outcomes of 609 cycles performed by FISH and 331 performed by array-CGH and NGS showed significant improvement of the application of next-generation technologies, resulting in almost doubling pregnancy rate, from 38.8 percent in FISH cycles to 66.5 percent with application of next-generation technologies, and twofold reduction of spontaneous abortion rate, from 18.1 percent to 8.9 percent.48 A few sophisticated approaches based on nextgeneration technologies have been developed for distinguishing noncarrier balanced embryos from normal ones. One such technology involved the use of an SNP microarray.166, 167 However, this method requires the availability of a lot of the unbalanced embryo, as well as parental DNA necessary to serve as a reference for distinguishing balanced translocation from normal blastocysts. The more universal approach is a specially designed NGS technology called mate-pair sequencing (MPS). This involves high-depth MPS to identify breakpoint regions and Sanger sequencing to define the exact breakpoint

needed for designing specific primers required to identify normal and carrier embryos.168 A similar approach, termed nanopore long-read sequencing, also discriminates carrier from noncarrier embryos through high-resolution breakpoint mapping followed by breakpoint PCR.169 Thus, following application of breakpoint PCR, carrier embryos can be discriminated from noncarrier embryos. Both these approaches enable accurate high-resolution breakpoint mapping directly on balanced reciprocal translocation carriers, providing the option of transferring euploid noncarrier embryos. Thus, the current technologies not only insure an acceptable pregnancy outcome for carriers of structural rearrangement, but also enable avoidance of balanced offspring and continuation of the problem in the next generation. The presented data provide strong evidence that PGT is currently an important alternative to prenatal diagnosis, as it widens the options available for couples wishing to avoid the birth of an affected child, and provides the possibility of having children for those who would remain childless because of their objection to termination of pregnancy following prenatal diagnosis. At the same time, PGT is also becoming an integral part of assisted reproduction, by avoiding transfer of chromosomally abnormal and potentially nonviable embryos, thereby contributing to a significant increase in implantation and pregnancy rates in

130 Genetic Disorders and the Fetus

IVF, and to a general improvement in the standards of assisted reproduction practices.

Ethical and legal issues Considerations on ethical and legal issues are evolving, along with the evolution of the technology for the control of genetic diseases, and have become one of the key subjects in discussing the acceptability of preconception and preimplantation testing for genetic disorders. Ethical and legal issues determine, to a considerable extent, whether these new approaches are promoted to become an integral part of preventive genetics services or are waived on ethical grounds.8 PGT could be regarded as an ethically acceptable procedure in the context of a general objective of genetic service, which, according to the WHO, is to help genetically disadvantaged people live and reproduce as normally and as responsibly as possible.9 Because PGT is heavily based on IVF, it also is relevant that IVF is considered to be ethically acceptable in many countries.170–174 However, complex ethical and legal issues are confronted differently in various countries (see Chapter 36).175 For example, in Germany, the future of PGT depends on an Embryo Protection Law, which has been in effect since 1991.172, 176 This law is very strict and prohibitive of embryo research. However, it prohibits only destructive research that impairs the chances of the embryo becoming a human being up to the eight-cell stage. In fact, blastocyst biopsy may be possible without any conflict with the law because, together with chorionic villus sampling (CVS), such embryo biopsies are considered beneficial, allowing decisions to be made before replacement. Therefore, there is no conflict about the provision of PGT in Germany before the pronucleate stage, which is currently under way. However, this must be done for diagnostic purposes only, not for research. Even in the case of tripronucleate embryos, only observation is permitted, not experimentation. This approach may also resolve the ethical issues impeding PGT in Austria, Switzerland, Malta, and other predominantly Catholic countries.177 The same holds for other countries where no preventive measures have been allowed on religious grounds. For example, the law has evolved recently in Switzerland, citizens voting to introduce PGT

into clinical practice. In contrast, in France, there seems to be no law at all concerning either PGT or embryo research. However, the attitude of the National Ethical Committee toward PGT is influenced by the fact that the diagnosis is based on genetic analysis of only one or a few cells and that all male embryos are discarded after gender determination, while half of them are completely normal. However, the testing is presently based on specific diagnosis, rather than gender determination. Another concern is that PGT increases the need for IVF, which is provided free of charge in France. Finally, prenatal diagnosis, also provided free of charge, was enough to avoid genetic disorder, so the provision of PGT was considered to be an additional prenatal test, without taking into account the suffering caused by selective abortions after prenatal diagnosis. Nevertheless, there are presently a few well-established centers in France providing a full range of PGT services. In Italy, PGT and many aspects of IVF were forbidden by Act of Parliament for almost 7 years. Only three oocytes were allowed to be aspirated for fertilization in vitro, clearly reflecting the opinions of the hierarchy of the Roman Catholic Church.178 Prior to the law, Italy had been among the most active centers involved in the development and application of PGT for genetic and chromosomal disorders. After the laws were enacted, it was difficult to perform PGT, which nevertheless is presently performed without restriction. In some countries, such as Belgium, the decision on embryo research and PGT rests completely with institutional review boards, so there is no problem with the development of the technique and its implementation into clinical practice. In other countries, such as the Netherlands, PGT is governed by the law on medical experiments, which contains a section on embryo research. It prohibits “cloning,” but probably will not ban PGT research because it provides an alternative to prenatal diagnosis and abortion of genetically affected fetuses. In the United Kingdom, PGT, as well as the practice of IVF and research involving human embryos, is regulated through a statutory body, the Human Fertilisation and Embryology Authority, and the Fertilisation and Embryology Act (1990), allowing research on human embryos up to 14 days of development under an appropriate license. In Spain, although a 1988 law regulating

CHAPTER 2

human embryo research forbade the fertilization of human oocytes for any purpose other than human procreation, it permitted research on embryos within 14 days of preimplantation development under the supervision of the national health and scientific authorities.179 Therefore, this law did not conflict with the development of research in preimplantation genetics and its application to assisted reproduction practices. In fact, according to a survey on PGT availability in Europe, the number of PGT centers presently available in the country is more than in most of the other European countries. In the United States and Australia, the legal status of PGT and community attitudes differ in different states. For example, in the six states of Australia, only three have laws governing IVF and embryo research. In Victoria, embryo research is prohibited, except for approved experiments, although this law does not actually affect PGT because IVF is allowed for infertile couples, and PGT also can be justified as the procedure for avoiding the risk of transmitting genetic disease to affected children. In Western Australia, PGT cannot be done because of the Experimentation Law, whereas in South Australia it is possible unless destructive to an implantable human embryo. In the United States, the issue of embryo research is closely associated with the debates on abortion and cloning, and there has been no government system for regulating reproductive research projects. Because there is no ethical advisory board (EAB) that is legally given responsibility for reviewing such research proposals, federal funding for human embryo research has not been available. In addition, a wide variation of policy positions exists among different states, mainly being compromised over consideration of the question of when human life begins. However, despite existing differences in current legal restrictions in this field, selection of pre-embryos on genetic grounds may be ethically acceptable based on the premise that the goal of avoiding the birth of offspring with severe genetic handicaps is part of the constitutional rights of procreative liberty.180, 181 Although the National Institutes of Health Revitalization Act of 1993 lifted the requirement (45 CFR 46.204.d) for a federal-level EAB review for IVF research, leaving consideration for clinical research relating to IVF to individual institutional review boards, none of

Preimplantation Genetic Testing 131

the federal funds may be used for research in which a human embryo or embryos are destroyed, discarded, or knowingly subjected to risk of injury or death greater than allowed for research on fetuses in utero. In Canada, recent legislation to regulate assisted human reproduction technologies has been introduced, entitled an “Act Respecting Assisted Human Reproduction,” which allows PGT for medical reasons but excludes identifying the sex of an embryo for social purposes.182, 183 The Society of Obstetricians and Gynaecologists of Canada have provided valuable guidelines that optimize obstetrical management and counseling for prospective parents undergoing IVF, integral to PGT. Emphasis is given to the increasing evidence that both infertility and subfertility remain as independent risk factors for subsequent complications and adverse perinatal outcomes, even without IVF. Their report also draws attention to the very low but actual risk of imprinting disorders, such as Beckwith–Wiedemann syndrome or Angelman syndrome, estimated to occur in fewer than one in 5,000 patients. Important ethical issues have recently been raised with increasing use of PGT for gender determination for social reasons,184, 185 late-onset disorders with genetic predisposition,6, 7, 186, 187 and PGT-HLA to produce an HLA-compatible donor to treat a family member with fatal bone marrow disease or cancer requiring a stem cell transplantation.5, 188, 189 Although there is no actual difference in the application of PGT for the latter conditions, the controversy can be explained by the fact that in traditional prenatal diagnosis, if the fetus was found to carry the gene predisposing to a late-onset disorder or to be HLA unmatched, a couple would have to make an extremely difficult decision about pregnancy termination, which could hardly be justified by such a finding. Alternatively, PGT technology allows genetic testing of human eggs and embryos before pregnancy is established, making it totally realistic to establish only HLA-matched or potentially normal pregnancies without genetic predisposition to late-onset disorders. Notwithstanding the foregoing considerations, PGT has become an established clinical option in reproductive medicine and is applied using separate consent forms and research protocols approved

132 Genetic Disorders and the Fetus

by institutional ethics committees. Despite recent controversy regarding the quantitation of the impact of PGT-A on clinical outcome, thousands of cases have been performed, resulting in the birth of thousands of healthy children born after PGT-A. However, these protocols still require confirmatory CVS or amniocentesis and follow-up monitoring of their safety and accuracy. Although PGT will help solve some of the longstanding ethical problems, such as the abortion issue (which could largely be avoided as a result of this approach), other issues could become a serious obstacle, particularly those related to “designer babies.” These considerations are highly relevant to the subject of PGT, as well as to any other new methods as we proceed with further development of appropriate technology for avoiding genetic disability.

Conclusion Although the introduction of first-trimester prenatal diagnosis by CVS has considerably improved the possibility of avoiding genetic diseases, selective abortion is an issue in the case of an affected fetus. PGT has been initiated to provide the option of avoiding the birth of an affected child without the need for abortion as an obligatory component in the prevention program. This chapter describes these important developments with the emphasis on addressing the problems of implementation of PGT into clinical practice. Currently, PGT has been applied clinically for up to 600 different conditions, with thousands of

unaffected children born after PGT performed for single-gene and chromosomal disorders. Among the approaches to PGT introduced, blastocyst biopsy is now a standard. This became possible due to the progress in micromanipulation and biopsy and genetic analysis of single cells or small number of cells by PCR and currently by next-generation technologies. The available experience has already demonstrated the practical utility of PGT, and the reliability and safety of this relatively new technology in assisted reproduction. The indications for PGT have been expanded beyond those used in prenatal diagnosis to include couples at high risk of having a child with a genetic disorder (in the face of antipathy toward elective abortion), poor-prognosis IVF patients, couples at risk for producing offspring with late-onset genetic disorders, and preimplantation HLA matching. Because of the high prevalence of chromosomal abnormalities in early pregnancy, the introduction of PGT-A will not only make it possible to avoid the risk of age-related aneuploidies, but will also considerably improve the embryo recovery and pregnancy outcome following PGT and should improve the effectiveness of IVF programs in general. Introduction of NGS-based PGT-A, which uses WGA as the first step of the technique, also makes it possible to perform PGT-M with or without PGT-HLA in the same biopsy material. Concomitant PGT-A with PGT-M, PGT-SR, and PGT-HLA are thus becoming standard procedures toward comprehensive PGT for genetic and chromosomal disorders.

References 1. Zegers-Hochschild F, David Adamson G, Dyer S, et al. The international glossary on infertility and fertility care. Fertil Steril 2017;108(3):393. 2. Verlinsky Y, Kuliev AM. Preimplantation genetics. New York: Plenum Press, 1991. 3. Verlinsky Y, Kuliev AM. Preimplantation diagnosis of genetic diseases: a new technique for assisted reproduction. New York: Wiley-Liss, 1993. 4. Verlinsky Y, Kuliev AMA. Atlas of preimplantation genetic diagnosis, New York: Parthenon, 2000. 5. Verlinsky Y, Rechitsky S, Schoolcraft W, et al. Preimplantation diagnosis for Fanconi anemia combined with HLA matching. JAMA 2001;285:3130.

6. Rechitsky S, Verlinsky O, Chistokhina A, et al. Preimplantation genetic diagnosis for cancer predisposition. Reprod Biomed Online 2002;5:148. 7. Verlinsky Y, Rechitsky S, Verlinsky O, et al. Preimplantation diagnosis for early onset Alzheimer disease caused by V717L mutation. JAMA 2002;287: 1038. 8. Kuliev AM, Modell B. Ethical issues in the control of genetic diseases. In: Verlinsky Y, Kuliev AM, eds. Preimplantation genetics. New York: Plenum Press, 1991:233. 9. World Health Organization. Perspectives of fetal diagnosis of congenital diseases. WHO unpublished

CHAPTER 2

10.

11. 12.

13.

14.

15.

16.

17.

18.

19.

20.

21.

22.

23.

24.

document HMG/Serono/84.4. Geneva: World Health Organization, 1984. Kuliev A, Modell B, Galjaard H. Perspectives in fetal diagnosis of congenital disorders. Arns Serono Symposia, Rome, 1985. McLaren A. Prenatal diagnosis before implantation: opportunities and problems. Prenat Diagn 1985;5:85. Saiki R, Scharf S, Faloona F, et al. Enzymatic amplification of beta-globin genomic sequences and restriction site analysis for diagnosis of sickle cell anemia. Science 1985;230:1350. Leonard M, Kirszenbaum M, Cotinot C, et al. Sexing bovine embryos using a Y chromosome specific probe. Theriogenology 1987;27:248. Monk M, Handyside AH. Sexing of preimplantation mouse embryos by measurement of X-linked gene dosage in single blastomere. J Reprod Fertil 1988;82:365. Monk M, Muggleton-Harris AL, Rawling E, et al. Preimplantation diagnosis of HPRT-deficient male mice and carrier female mouse embryo by trophectoderm biopsy. Hum Reprod 1988;3:377. Summers PM, Campbell JM, Miller MW. Normal in-vivo development of marmoset monkey embryos after trophectoderm biopsy. Hum Reprod 1988;3: 389. Gordon JW, Gang I. Use of zona drilling for safe and effective biopsy of murine oocytes and embryos. Biol Reprod 1990;42:869. Handyside AH, Pattison JK, Penketh RJA, et al. Biopsy of human preimplantation embryos and sexing by DNA amplification. Lancet 1989;1:347. Handyside AH, Kontogiani EH, Hardy K, et al. Pregnancies from biopsied human preimplantation embryos sexed by Y-specific DNA amplification. Nature 1990;344:768. Verlinsky Y, Ginsberg N, Lifchez A, et al. Analysis of the first polar body: preconception genetic diagnosis. Hum Reprod 1990;5:826. Verlinsky Y, Cohen J, Munne S, et al. Over a decade of preimplantation genetic diagnosis experience – a multicenter report. Fertil Steril 2004;82:292. ESHRE Preimplantation Genetic Diagnosis (PGD) Consortium. Data collection X. Hum Reprod 2010;25:2685. Liebaers I, Desmyttere S, Verpoest W, et al. Report on a consecutive series of 581 children born after blastomere biopsy for preimplantation genetic diagnosis. Hum Reprod 2010;25:275. Preimplantation Genetic Diagnosis International Society (PGDIS). 12th International Conference on Preimplantation Genetic Diagnosis. Reprod BioMed Online 2013;26:Suppl 1.

Preimplantation Genetic Testing 133

25. Kuliev A. Practical preimplantation genetic diagnosis. New York, London, Heidelberg: Springer, 2013. 26. Preimplantation Genetic Diagnosis International Society (PGDIS). Guidelines for good practice in PGD: program requirements and laboratory quality assurance. 2008. Reprod BioMed Online 2008;16:134. 27. ESHRE Preimplantation Genetic Diagnosis (PGD) Consortium. Best practice guidelines for preimplantation genetic diagnosis/screening (PGD/PGS). Hum Reprod 2011;26:14. 28. Kuliev A, Rechitsky S, Verlinsky O, et al. Preembryonic diagnosis for sickle cell disease. Mol Cell Endocrinol 2001;183:S19. 29. Lacham-Kaplan O, Daniels R, Trouson A. Fertilization of mouse oocytes using somatic cells as male germ cells. Reprod Biomed Online 2001;3:205. 30. Tesarik J, Mendoza C. Somatic cell haploidization: an update. Reprod BioMed Online 2003;6:60. 31. Willadsen S, Munne S, Schmmel T, et al. Applicationsof nuclear sperm duplication. Fifth International Symposium on Preimplantation Genetics, 5–7 June, Antalya, Turkey 2003:35. 32. Kuznyetsov V, Kuznyetsova I, Chmura M, et al. Duplication of sperm genome by human androgenetic embryo production for possible testing of paternal genome prior to fertilization. Reprod BioMed Online 2007;14:504. 33. Modlinsky J, McLaren A. A method for visualizing the chromosomes of the second polar body of the mouse egg. J Embryol Exp Morphol 1980;60:97. 34. Monk M, Holding C. Amplification of beta-haemoglobin sequence in individual human oocytes and polar bodies. Lancet 1990;335:985. 35. Kuliev A, Rechitsky S. Polar body-based preimplantation genetic diagnosis for Mendelian disorders. Mol Hum Reprod 2011;17:275. 36. Kaplan B, Wolf G, Kovalinskaya L, et al. Viability of embryos following second polar body removal in a mouse model. Assist Reprod Genet 1995;12:747. 37. Hardy K, Handyside AH, Winston RML. The human blastocyst: cell number, death and allocation during late preimplantation development in vitro. Development 1989;107:597. 38. Gardner RL, Edwards RG. Control of the sex ratio at full term in the rabbit by transferring sexed blastocysts. Nature 1968;218:346. 39. Dokras A, Sargent IK, Ross C, et al. Trophectoderm biopsy in human blastocyst. Hum Reprod 1990;5:821. 40. Dokras A, Sargent IK, Ross C, et al. The human blastocyst: morphology and human chorionic gonadotrophin secretion in vitro. Hum Reprod 1991;6:1143.

134 Genetic Disorders and the Fetus

41. Muggleton-Harris AL, Glazier AM, Pickering S, et al. Genetic diagnosis using PCR and FISH analysis of biopsied cells from both the cleavage and blastocyst stages of individual cultured human preimplantation embryos. Hum Reprod 1995;10:183. 42. Verlinsky Y, Munne S, Simpson JL, et al. Current status of preimplantation diagnosis. J Assist Reprod Genet 1997;1:72. 43. Dawson KJ, Rutherford AJ, Winston NJ, et al. Human blastocyst transfer: is it a feasible proposition? Hum Reprod 1988;145:44. 44. McArthur SJ, Leigh D, Marshall JT, et al. Pregnancies and live births after trophectoderm biopsy and preimplantation genetic testing of human blastocysts. Fertil Steril 2005;84:1628. 45. De Boer KA, Catt JW, Jansen RPS, et al. Moving to blastocyst biopsy for preimplantation genetic diagnosis and single embryo transfer at Sydney IVF. Fertil Steril 2004;2:295. 46. PGDIS. Eighth International Symposium on Preimplantation Genetic Diagnosis. Reprod BioMed Online 2008;16:60. 47. Scott R, Upham KM, Forman EJ, et al. Cleavage-stage biopsy significantly impairs human embryonic implantation potential while blastocyst biopsy does not: a randomized and paired clinical trial. Fertil Steril 2013;10:624. 48. Kuliev A, Rechitsky S, Simpson JL. Practical preimplantation genetic testing. New York: Springer, 2020. 49. Gianaroli L, Magli MC, Pomante A, et al. Blastocentesis: a source of DNA for preimplantation genetic testing. Results from a pilot study. Fertil Steril 2014;102:1692. 50. Magli C, Andor C, Tabanelli C, et al. Deoxyribonucleic acid detection in blastocoelic fluid: a new predictor of embryo ploidy and viable pregnancy. Fertil Steril 2019;111:77. 51. Shamonki MI, Jin H, Haimowitz Z, Liu L. Proof of concept: preimplantation genetic screening without embryo biopsy through analysis of cell-free DNA in spent embryo culture media. Fertil Steril 2016;106:1312. 52. Xu J, Fang R, Chen L, et al. Noninvasive chromosome screening of human embryos by genome sequencing of embryo culture medium for in vitro fertilization. Proc Natl Acad Sci U S A 2016;113:11907. 53. Hammond ER, McGillivray BC, Wicker SM, et al. Characterizing nuclear and mitochondrial DA in spent embryo culture media: genetic contamination identified. Fertil Steril 2017;107:220. 54. Liu W, Liu J, Du H, et al. Non-invasive preimplantation aneuploidy screening and diagnosis

55.

56.

57.

58.

59. 60.

61.

62.

63.

64. 65.

66.

67.

68.

of beta thalassemia IVSII654 mutation using spent embryo culture medium. Ann Med 2017;49(4):319. Katz-Jaffe M, McCallie B, McReynolds S, et al. Analysis of the embryonic media drop as a non-invasive alternative to blastocyst biopsy in preimplantation genetic testing for aneuploidy. Fertil Steril 2018;110(4):Suppl e419. Capablo A, Romanelli V, Patassini C, et al, Diagnostic accuracy of blastocoelic fluid and spent media as source of DNA for preimplantation genetic testing in standard clinical conditions. Fertil Steril 2018;110: 870. Ho JR, Arrach N, Rhodes K, et al. Pushing the limits of detection: investigation of cell-free DNA for aneuploidy screening in embryos. Fertil Steril 2018;110: 467. Rubio C. Non-Invasive PGT-A with the analysis of embryonic cell free DNA in spent blastocyst media. PGDIS 2019, Reprod Biomed Online 2019;39:E12. Verlinsky Y, Kuliev A. Atlas of preimplantation genetic diagnosis. London: Taylor and Francis, 2005:288. Rechitsky S, Strom C, Verlinsky O, et al. Allele drop out in polar bodies and blastomeres. J Assist Reprod Genet 1998;15:253. Simpson JL, Rechitsky S, Kuliev A. Next generation sequencing for preimplantation genetic diagnosis. Fertil Steril 2013;99:1203. Renwick P, Trussler J, Braude P, et al. Preimplantation genetic haplotyping: 127 diagnostic cycles demonstrating a robust, efficient alternative to direct mutation testing on single cells. Reprod BioMed Online 2010;20:470. Verlinsky Y, Rechitsky S, Verlinsky O, et al. Preimplantation genetic diagnosis for polycystic kidney disease. Fertil Steril 2004;82:926. Verlinsky Y, Kuliev A. Practical preimplantation genetic diagnosis. Berlin: Springer, 2006:204. Handyside AH, Thornhill AR, Harton GL, et al. Karyomapping: a novel molecular karyotyping method based on mapping crossovers between parental haplotypes with broad applications for preimplantation genetic diagnosis of inherited disease. J Med Genet 2010;47:651. Verlinsky Y, Rechitsky S, Verlinsky O, et al. Polar body based preimplantation diagnosis for X-linked genetic disorders. Reprod BioMed Online 2002;4:38. Kuliev A, Rechitsky S, Laziuk K, et al. Pre-embryonic diagnosis for Sandhoff disease. Reprod BioMed Online 2006;12:328. Verlinsky Y, Milayeva S, Evsikov S, et al. Preconception and preimplantation diagnosis for cystic fibrosis. Prenat Diagn 1992;12:103.

CHAPTER 2

69. Munne S, Morrison L, Fung J, et al. Spontaneous abortions are reduced after preconception diagnosis of translocations. J Assist Reprod Genet 1998;15: 290. 70. Montag M, van der Ven K, Dorn C, et al. Outcome of laser-assisted polar body biopsy and aneuploidy testing. Reprod Bio Med Online 2004;9:425. 71. Magli C, Ferraretti A, Crippa A, et al. First meiosis errors in immature oocytes generated by stimulated cycles. Fertil Steril 2006;86:629. 72. Amato P, Tashibana M, Sparman M, et al. Three-patient in vitro fertilization: gene replacement for the prevention of inherited mitochondrial diseases. Fert Steril 2014;101:31. 73. Rechitsky S, Pomerantseva K, Pakhalchuk T, et al. First systematic experience of preimplantation genetic diagnosis for de novo mutations. Reprod BioMed Online 2011;22:350. 74. Verlinsky Y, Rechitsky S, Verlinsky O, et al. Preimplantation diagnosis for p53 tumor suppressor gene mutations. Reprod Bio Med Online 2001;2:102. 75. Verlinsky Y, Rechitsky S, Verlinsky O, et al. Preimplantation diagnosis for sonic hedgehog mutation causing familial holoprosencephaly. N Engl J Med 2003;348:1449. 76. Verlinsky Y, Rechitsky S, Schoolcraft, et al. Preimplantation diagnosis for homeobox gene HLXB9 mutation causing Currarino syndrome. Am J Med Genet 2005;134A:103. 77. He J, McDermont DA, Song Y, et al. Preimplantation genetic diagnosis of human congenital heart disease and Holt–Oram syndrome. Am J Med Genet 2003;126A:93. 78. Kuliev A, Pomerantseva E, Polling D, et al. PGD for inherited cardiac diseases. Reprod Bio Med Online 2012;24:443 79. Verlinsky Y, Rechitsky S, Sharapova T, et al. Preimplantation HLA typing. JAMA 2004;291:2079. 80. Edwards RG. Ethics of PGD: thoughts on the consequences of typing HLA in embryos. Reprod Bio Med Online 2004 9:222. 81. Rechitsky S, Kuliev A, Tur-Kaspa I, et al. Preimplantation HLA typing with preimplantation genetic diagnosis. Reprod Bio Med Online 2004;6:488. 82. vande Velde H, Georgiou I, de Rycke M, et al. Novel universal approach for preimplantation genetic diagnosis of β-thalassemia in combination with HLA matching of embryos. Hum Reprod 2004;19:700. 83. Kahraman S, Karlilaya G, Sertyel S, et al. Clinical aspects of preimplantation genetic diagnosis of single gene disorders combined with HLA typing. Reprod BioMed Online 2004;9:529.

Preimplantation Genetic Testing 135

84. Kuliev A, Rechitsky S, Verlinsky O, et al. Preimplantation diagnosis and HLA typing for hemoglobin disorders. Reprod BioMed Online 2005;11:362. 85. Rechitsky S, Kuliev A, Sharapova T, et al. Preimplantation HLA typing with aneuploidy testing. Reprod BioMed Online 2006;12:81. 86. Kuliev A, Rechitsky S, Tur-Kaspa I, et al. Preimplantation genetics: improving access to stem cell therapy. Ann NY Acad Sci 2005;1054:223. 87. Chen SU, Su YN, Fang MY, et al. PGD of betathalassaemia and HLA haplotypes using OmniPlex whole genome amplification. Reprod Biomed Online 2008;17:699. 88. Verlinsky Y, Rechitsky S, Sharapova T, et al. Preimplantation diagnosis for immunodeficiencies. Reprod Biomed Online 2007;14:214–223. 89. Reichenbach J, van de Velde H, de Rycke M, et al. First successful bone marrow transplantation for X-linked chronic granulomatous disease by using preimplantation female gender typing and HLA matching. Pediatrics 2008;122:e778. 90. Goussetis E, Kokkali G, Petrakou E, et al. Successful hematopoietic stem cell transplantation in 2 children with X-linked chronic granulomatous disease from their unaffected HLA-identical siblings selected using preimplantation genetic diagnosis combined with HLA typing. Biol Blood Marrow Transplant 2010;16:344. 91. Kuliev A, Packalchuk T, Verlinsky O, et al. Preimplantation diagnosis: efficient tool for human leukocyte antigen matched bone marrow transplantation for thalassemia. Thalassemia Rep 2011;1:e1. 92. Kuliev A, Rechitsky S, Verlinsky O. Atlas of preimplantation genetic diagnosis, 3rd edn. London: CRS Press, Taylor and Francis, 2014. 93. Kahraman S, Beyazyurek C, Ekmeksi C, et al. Seven year experience of preimplantation HLA typing: a clinical experience of 327 cycles. Reprod Biomed Online 2011;23:363. 94. Umay KB, Gavaz M, Kumtepe ÇY, et al. Successful hemapoietic stem cell transplantation in 62 children from healthy siblings conceived from preimplantation HLA Matching: a clinical experience of 327 cycles. Reprod Biomed Online 2019;39(Suppl 1):e13. 95. Plachot M. Chromosomal abnormalities in oocytes. Mol Cell Endocrinol 2001;183:S59. 96. Martin RH, Spriggs E, Rademaker AW. Multicolour fluorescence in situ hybridization analysis of aneuploidy and diploidy frequencies in 225,846 sperm from ten normal men. Biol Reprod 1996;54:394. 97. Gropp A. Chromosomal animal model of human disease: fetal trisomy and development failure. In: Berry L,

136 Genetic Disorders and the Fetus

98.

99.

100.

101.

102.

103.

104. 105.

106.

107.

108.

109.

110.

111.

Poswillo DE, eds. Teratology. Berlin: Springer-Verlag, 1975:17. Epstein CJ. Mouse monosomies and trisomies as experimental systems for studying mammalian aneuploidy. Trends Genet 1985;1:129. Dyban A, De Sutter P, Verlinsky Y. Preimplantation cytogenetic analysis. In: Verlinsky Y, Kuliev AM, eds. Preimplantation diagnosis of genetic diseases: a new technique in assisted reproduction. New York: Wiley-Liss, 1993:93. De Sutter P, Dozortsev D, Cieslak J, et al. Parthenogenetic activation of human oocytes by puromycin. J Assist Reprod Genet 1992;9:328. Dyban AP, de Sutter P, Dozortsev D, et al. Visualization of second polar body chromosomes in fertilized and artificially activated mouse oocytes treated with okadaic acid. J Assist Reprod Genet 1992;9:572. Verlinsky Y, Dozortsev D, Evsikov S. Visualization and cytogenetic analysis of second polar body chromosomes following its fusion with one-cell mouse embryo. J Assist Reprod Genet 1994;11:123. Antonarakis SE, Petersen MB, McInnis MG, et al. The meiotic stage of nondisjunction in trisomy 21: determination by using DNA polymorphisms. Am J Hum Genet 1992;50:544. Rodman TC. Chromosomes of the first polar body in mammalian meiosis. Exp Cell Res 1971;68:205. Dyban A, de Sutter P, Verlinsky Y. Okadaic acid induces premature chromosome condensation reflecting the cell cycle progression in one cell stage mouse embryos. Mol Reprod Dev 1993;34:403. Tanaka A, Nagayoshi M, Awata S, et al. Preimplantation diagnosis of repeated miscarriage due to chromosomal translocations using metaphase chromosomes of a blastomere biopsied from 4–6 cell stage embryo. Fertil Steril 2004;81:30. Dyban A, Fredine M, Severova E, et al. Detection of aneuploidy in human oocytes and corresponding first polar bodies by FISH. J Assist Reprod Genet 1995;13:72. Munné S, Dailey T, Sultan KM, et al. The use of first polar bodies for preimplantation diagnosis of aneuploidy. Hum Reprod 1995;10:1014. Verlinsky Y, Cieslak J, Freidine M, et al. Polar body diagnosis of common aneuploidies by FISH. J Assist Reprod Genet 1996;13:157. Verlinsky Y, Cieslak J, Ivakhnenko V, et al. Birth of healthy children following preimplantation diagnosis of common aneuploidies by polar FISH analysis. Fertil Steril 1996;66:126. Kuliev A, Cieslak J, Illkewitch Y, et al. Chromosomal abnormalities in a series of 6733 human oocytes in

112.

113.

114.

115.

116.

117.

118.

119.

120.

121.

122.

123.

124.

125.

preimplantation diagnosis of age-related aneuploidies. Reprod Biomed Online 2003 6:54. Kuliev A, Cieslak J, Verlinsky Y. Frequency and distribution of chromosomal abnormalities in human oocytes. Cytogenet Genome Res 2005;111:193. Kuliev A, Zlatopolsky Z, Kirillova I, et al. Meiosis errors in over 20,000 oocytes studied in the practice of preimplantation aneuploidy testing. Reprod BioMed Online 2011;22:2. Munne S. Preimplantation genetic diagnosis of numerical and structural chromosome abnormalities. Reprod Biomed Online 2002;4:183. Gianaroli L, Magli MC, Ferraretti AP. The in vivo and in vitro efficiency and efficacy of PGD for aneuploidy. Mol Cell Endocrinol 2001;183:S13. Munné S, Chen S, Colls P, et al. Maternal age, morphology, development and chromosome abnormalities in over 6000 cleavage stage embryos. Reprod BioMed Online 2007;14:628. Colls P, Escudero T, Cekleniak N, Sadowy S, Cohen J, Munne S. Increased efficiency of preimplantation genetic diagnosis for aneuploidy by testing 12 chromosomes. Reprod Biomed Online 2009;19:532. Uher P, Baborova P, Kralickova M, et al. Noninformative results and monosomies in PGD: the importance of a third round of re-hybridization. Reprod Biomed Online 2009;18:530. Fragouli E, Alfarawati S, Spath K, The origin and impact of embryonic aneuploidy. Hum Genet 2013;132:1001. Munné S, Sandalinas M, Escudero T, et al. Improved implantation after preimplantation genetic diagnosis of aneuploidy. Reprod BioMed Online 2003;7:91. Munné S, Fisher J, Warner A, et al. Preimplantation genetic diagnosis significantly reduces pregnancy loss in infertile couples: a multi-center study. Fertil Steril 2006;85:326. Staessen C, Platteau P, van Assche E, et al. Comparison of blastocyst transfer with or without preimplantation genetic diagnosis for aneuploidy in couples with advanced maternal age: a prospective randomized controlled trial. Hum Reprod 2004;19:2849. Platteau P, Staessen C, Michiels A, et al. Preimplantation genetic diagnosis for aneuploidy in patients with unexplained recurrent miscarriages. Fertil Steril 2005;83:393. Mastenbroek S, Twisk M, van Echten-Arends J, et al. In vitro fertilization with preimplantation genetic screening. N Engl J Med 2007;357:9. Cohen J, Wells D, Munné S. Removal of two cells from cleavage stage embryos is likely to reduce the efficacy of

CHAPTER 2

126.

127.

128.

129.

130.

131.

132.

133.

134.

135.

136.

137.

138.

chromosomal tests employed to enhance implantation rates. Fertil Steril 2007;87:496. Munné S, Cohen J, Simpson JL. In vitro fertilization with preimplantation genetic screening. N Engl J Med 2007;357:1769. Cohen J, Grifo J. Multicentre trial of preimplantation genetic screening reported in the New England Journal of Medicine: an in-depth look at the findings. Reprod Biomed Online 2008;15:365. Practice Committee of the Society for Assisted Reproductive Technology and Practice Committee of the American Society for Reproductive Medicine. Preimplantation genetic testing: a Practice Committee opinion. Fertil Steril 2007;88:1497. Lathi RB, Westphal LM, Milki AA. Aneuploidy in the miscarriages of infertile women and the potential benefit of preimplantation genetic diagnosis. Fertil Steril 2008;89:353. Gianaroli L, Magli MC, Ferraretti A. The beneficial effects of PGD for aneuploidy support extensive clinical application. Reprod BioMed Online 2004;10: 633. Verlinsky Y, Tur-Kaspa I, Cieslak J, et al. Preimplantation testing for chromosomal disorders improves reproductive outcome of poor-prognosis IVF patients. Reprod BioMed Online 2005;11:219. Magli MC, Jones GM, Gras L, et al. Chromosome mosaicism in day 3 aneuploid embryos that develop to morphologically normal blastocysts in vitro. Hum Reprod 2000;15:1781. Sandalinas M, Sadowy S, Alikani M, et al. Developmental ability of chromosomally abnormal human embryos to develop to the blastocyst stage. Hum Reprod 2001;16:1954. Ankum WM, Reitsma JB, Offringa M. IVF with preimplantation genetic screening, a promising new treatment with unexpectedly negative health outcomes: the Hippocratic role of Data Monitoring Committees. Hum Reprod 2008;23:1. Twisk M, Mastenbroek S, Hoek A. No beneficial effect of preimplantation genetic screening in women of advanced maternal age with a high risk for embryonic aneuploidy. Hum Reprod 2008;23:2813. Yakin K, Urman B. What next for preimplantation genetic screening? A clinician’s perspective. Hum Reprod 2008;23:1686. Gleicher N, Kushnir V, Barad D. Preimplantation genetic screening: still in search of a clinical application: a systematic review. Reprod Biol Endocrin 2014;12:22. Yang Z, Liu J, Collinz GS, et al. Selection of single blastocysts for fresh transfer via standard morphology

139.

140.

141.

142.

143.

144.

145.

146.

147.

148.

149.

Preimplantation Genetic Testing 137

assessment alone and with array CGH for good prognosis IVF patients: results from a randomized pilot study. Mol Cytogenet 2012;5:24. Scott RT, Tao X, Ferry KM, Treff NR. A prospective randomized controlled trial demonstrating significantly increased clinical pregnancy rates following 24 chromosome aneuploidy screening: biopsy and analysis on day 5 with fresh transfer. Fertil Steril 2010;94(Suppl):S2. Forman EJ, Tao X, Ferry KM, et al. Single embryo transfer with comprehensive chromosome screening results in improved ongoing pregnancy rates and decreased miscarriage rates. Hum Reprod 2012;27:1217. Rubio C, Bellver J, Rodrigo L, et al. In vitro fertilization with preimplantation genetic diagnosis for aneuploidies in advanced maternal age: a randomized, controlled study. Fertil Steril 2017;107:1122. Society for Assisted Reproductive Technology. SART national summary report: Final CSR for 2016. https://www.sartcorsonline.com/rpt\ignorespacesSR_ PublicMultYear.aspx? (accessed May 6, 2019). Munné S, Kaplan B, Frattarelli JL, et al.; STAR Study Group. Preimplantation genetic testing for aneuploidy versus morphology as selection criteria for single frozen-thawed embryo transfer in good-prognosis patients: a multicenter randomized clinical trial. Fertil Steril 2019;112(6):1071. Schoolcraft WB, Fragouli E, Stevens J, et al. Clinical application of comprehensive chromosomal screening at the blastocyst stage. Fertil Steril 2010;94: 1700. Fragouli E, Alfarawati S, Daphnis DD, et al. Cytogenetic analysis of human blastocyst with the use of FISH, CGH, and a CGH: scientific data and technical evaluation. Hum Reprod 2011;26:480. Schoolcraft WB, Treff NR, Stevens JM, et al. Live birth outcome with tro-phectoderm biopsy, blastocyst vitrification, and single-nucleotide polymorphism microarray-based comprehensive chromosome screening in infertile patients. Fertil Steril 2011;96:638. Geraedts J, Montag M, Magli C, et al. Polar body array CGH for prediction of the status of the corresponding oocyte. Part I: clinical results. Hum Reprod 2011;26:3172. Magli C, Montag M, Koster M, et al. Polar body array CGH for prediction of the status of the corresponding oocyte. Part II: technical aspects. Hum Reprod 2011;26(11):3181. Gabriel AS, Thornhill AR, Ottolini CS, et al. Array comparative genomic hybridization on first polar bodies suggests that non-disjunction is not the

138 Genetic Disorders and the Fetus

150.

151.

152.

153.

154.

155.

156.

157.

158.

159.

160.

161.

predominant mechanism leading to aneuploidy in humans. J Med Genet 2011;48:433. Brezina PR, Benner A, Rechitsky S, et al. Single-gene testing combined with single nucleotide polymorphism microarray preimplantation genetic diagnosis for aneuploidy: a novel approach in optimizing pregnancy outcome. Fertil Steril 2011;95:1786. Treff NR, Northrop LE, Kasabwala K, et al. Single nucleotide polymorphism microarray-based concurrent screening of 24-chromosome aneuploidy and unbalanced translocations in preimplantation human embryos. Fertil Steril 2011;95:1606. Scott RT Jr., Ferry K, Su J, et al. Comprehensive chromosome screening is highly predictive of the reproductive potential of human embryos: a prospective, blinded, nonselection study. Fertil Steril 2012;97:870. Colls P, Escudero T, Fleicher J, et al. Validation of array comparative genome hybridization for diagnosis of translocations in preimplantation human embryos. Reprod BioMed Online 2012;24:621. Munne S. PGD for aneuploidy and translocations using array comparative genome hybridization. Curr Genomics 2012;13:463. Rubio C, Rodrigo L, Mir P, et al. Use of array comparative genomic hybridization (array-CGH) for embryo assessment: clinical results. Fertil Steril 2013;99:1044. Harton GL, Munné S, Surrey M, et al. Diminished effect of maternal age on implantation after preimplantation genetic diagnosis with array comparative genomic hybridization. Fertil Steril 2013;100:1695. Rechitsky S, Verlinsky O, Kuliev A. PGD for cystic fibrosis patients and couples at risk of an additional genetic disorder combined with 24-chromsome aneuploidy testing. Reprod BioMed Online 2013;26: 420. Treff N, Scott RT. Four-hour quantitative real-time polymerase chain reaction-based comprehensive chromosome screening and accumulating evidence of accuracy, safety, predictive value, and clinical efficiency. Fertil Steril 2013;99:1049. Treff N, Fedic A, Xin T, et al. Evaluation of targeted next-generation sequencing-based preimplantation genetic diagnosis of monogenic disorders. Fertil Steril 2013;99:1377. Martin J, Cervero A, Mir P, et al. The impact of next generation sequencing technology on preimplantation genetic diagnosis and screening. Fertil Steril 2013;99:1054. Yin X, Tan K, Vajta G, et al. Massively parallel sequencing for chromosomal abnormality testing in trophectoderm cells of human blastocysts. Biol Reprod 2013;88:1.

162. Wells D, Kaur K, Grifo J, et al. A novel embryo screening provides new insights into embryo biology and yields the first pregnancies following genome sequencing. Hum Reprod 2013;28(Suppl 1):i25. 163. Fiorentino F, Biricik A, Bono S, et al. Development and validation of a next-generation sequencing-based protocol for 24-chromosome aneuploidy screening of embryos. Fertil Steril 2014;101(5):1375. 164. PGDIS Position statement on chromosome mosaicism and preimplantation aneuploidy testing at the blastocyst stage, 2016. PGDIS Newsletter, July 19, 2016 (www .pgdis.org). 165. PGDIS Position statement on transfer of mosaic embryos in preimplantation genetic testing for aneuploidy. Reprod Biomed Online 2019;39:e1. 166. Treff NR, Tao X, Schileings W, et al. Use of single nucleotide polymorphism microarrays to distinguish between balanced and normal chromosomes in embryos from a translocation carrier. Fertil Steril 2011;96:e58. 167. Treff NR, Thompson K, Rafizadeh M, et al. SNP array-based analysis of unbalanced embryos as a reference to distinguish between balanced translocation carriers and normal blastocysts. J Assist Reprod Genet 2016;38:1115. 168. Kuliev A, Zlatopolsky Z, Wang L, et al. Evolution of PGD for translocations. Abstracts of 15th International Conference on Preimplantation Genetics, Bologna, Italy, 2016. 169. Chow JFC, Cheng HH, Lau EYL, et al. Selective transfer of euploid non-carrier embryos with the use of long-read sequencing in preimplantation genetic testing for reciprocal translocation. Reprod Biomed Online 2019;39(Suppl 1):e14. 170. Ethics Committee of the American Fertility Society. Ethics and the new reproductive technologies. Fertil Steril 1990;53:5. 171. Walters L. Ethics and new reproductive technologies: an international review of committee statements. Hastings Cent Rep 1987;17(3). 172. Milunsky A. Ethical and selected medical aspects of preimplantation genetic diagnosis. In: Verlinsky Y, Kuliev AM, eds. Preimplantation genetics. New York: Plenum Press, 1991:245. 173. Cohen J, Hotz RL. Human embryo research: ethics and recent progress. Curr Opin Obstet Gynecol 1991;3:678. 174. Burn J, Strachan T. Human embryo use in developmental research. Nat Genet 1995;11:3. 175. Verlinsky Y, Handyside AH, Simpson JL. Current progress in preimplantation genetic diagnosis. J Assist Reprod Genet 1993;10:353.

CHAPTER 2

176. Schreiber HL. The legal situation regarding assisted reproduction in Germany. Reprod Biomed Online 2003;6:8. 177. Feichtinger W. Preimplantation diagnosis – a European clinician’s point of view. J Assist Reprod Genet 2004;21:15. 178. Benagiano P, Gianaroli L. The new Italian IVF legislation. Reprod Biomed Online 2004;9:117. 179. Peinado JA, Russell SE. The Spanish law governing assisted reproduction techniques: a summary. Hum Reprod 1990;5:634. 180. Robertson J. Extending preimplantation genetic diagnosis: the ethical debate. Ethical issues in new uses of preimplantation genetic diagnosis. Hum Reprod 2003;18:465. 181. Marshal E. Embryologists dismayed by sanctions against geneticist. Science 1997;275:472. 182. Gali RP, Woodside JL. Proposed Canadian legislation to regulate reproductive technologies and related research. Reprod Biomed Online 2003;6:114. 183. Society of Obstetricians and Gynaecologists of Canada, Okun N, Sierra S. Pregnancy outcomes after

184.

185.

186.

187.

188.

189.

Preimplantation Genetic Testing 139

assisted human reproduction. J Obstet Gynaecol Can 2014;36:64. Kilani Z, Haj Hassan L. Sex selection and preimplantation genetic diagnosis at the Farah Hospital. Reprod Biomed Online 2002;4:8. Malpani A, Malpani A, Modi D. The use of preimplantation genetic diagnosis in sex selection for family balancing in India. Reprod Biomed Online 2001;4:16. Ethics Committee of the American Society for Reproductive Medicine. Use of preimplantation genetic diagnosis for serious adult onset conditions: a committee opinion. Fertil Steril 2013;100:54. Edwards RG, Angastiniotis M, Antinoty S, et al. Ethics of preimplantation diagnosis: record from the Fourth International Symposiumon Preimplantation Genetics. Reprod Biomed 2003;6:170. Towner D, Loewy RS. Ethics of preimplantation diagnosis for a woman destined to develop early-onset Alzheimer disease. JAMA 2002;287:1038. Damewood MD. Ethical implications of a new application of preimplantation diagnosis. JAMA 2001;285:3143.

3

Amniotic Fluid Constituents, Cell Culture, and Neural Tube Defects Daniel L. Van Dyke 1 and Aubrey Milunsky 2,3 1

Mayo Medical School and Mayo Clinic Cytogenetics Laboratory, Rochester, MN, USA Center for Human Genetics, Cambridge, MA, USA 3 Tufts University School of Medicine, Boston, MA, USA 2

Introduction Amniotic fluid (AF) represents a constantly changing environment that simultaneously reflects and contributes to fetal development. Constituents include growth-promoting and growth-protective factors, and sufficient AF volume (AFV) provides mechanical cushioning and space for fetal movement. Biochemical and molecular components may also reflect fetal disease and maturity and, on occasion, maternal disease or environmental exposures. Analysis of the chemical constituents of AF has yielded valuable information for prenatal diagnosis, allowing assessment of fetal physiology and metabolism. Because the AF can be viewed as an extension of the fetal extracellular space,1, 2 an understanding of its origin, formation, and chemical constitution is crucial to prenatal diagnosis and fetal therapy. Sampling of extracoelomic fluid and AF during the 8th–16th weeks of pregnancy for the purpose of prenatal diagnosis has added valuable knowledge about the origin, formation, and content of AF.

Amniotic fluid Formation and circulation

Fluid exchange between the fetus and the mother occurs via several routes and through different mechanisms, and varies throughout pregnancy. Large volumes of fluid are transferred across the

fetal membranes, which are made up of five layers of amnion and four layers of chorion.3 Electron microscopy of the amnion has revealed a complex system of tiny intracellular canals that are connected to the intercellular canalicular system and the base of the cell.4 Studies in primates suggest that the AF is a transudate of the maternal plasma and becomes like other fetal fluids in the presence of the fetus, which contributes urine and other body secretions to the AF.5 Osmotic or diffusion permeability, hydrostatic pressure, chemical gradients, and other mechanisms are responsible for the fluid exchange between fetus and mother.6 In normal pregnancies, intra-amniotic pressure at 16 weeks ranges between 1 and 14 mmHg.7 Fisk et al.8 studied AF pressure from 7 to 38 weeks and found that it increased with gestational age and may be determined by anatomic and hormonal influences or gravid uterine musculature, but was not influenced by the deepest vertical pool, AF index, maternal age, parity, gravity, fetal sex, twinning, or time of delivery. These authors suggested that AF pressure did not change significantly after removing fluid samples in early or late amniocentesis.9 During the second trimester, total AF turnover is complete within about 3 hours.10 About 20 mL of AF/hour is swallowed by the fetus; that is, approximately 500 mL/day.11 At term, the exchange rate between fetus and mother may approach 500 mL/h.10, 12

Genetic Disorders and the Fetus: Diagnosis, Prevention, and Treatment, Eighth Edition. Edited by Aubrey Milunsky and Jeff M. Milunsky. © 2021 Aubrey Milunsky and Jeff M. Milunsky. Published 2021 by John Wiley & Sons Ltd.

140

CHAPTER 3

Amniotic Fluid Constituents, Cell Culture, and Neural Tube Defects 141

Although the fetus depends largely on the placenta for nutrient transport, it is also protected from marked fluctuations in maternal metabolism. The increase of creatinine, 𝛼-glutamyl transferase, and 𝛽 2 -microglobulin concentrations in AF after 10 weeks confirms the maturation of fetal glomerular function and reflects the fetal kidney development from the mesonephros to the metanephros.13 Active renal function is evident from the ability of the fetal kidney to concentrate radiopaque substances given intravenously to the mother, thereby allowing visualization of a fetal pyelogram.14 AF is mainly produced by the fetal kidney as pregnancy progresses and oligohydramnios may reflect renal structural anomalies, impaired swallowing, placental pathology, or general growth restriction.15 Volume

Brace16 described three determinants of AFV: (i) movement of water and solutes across the membranes; (ii) physiologic regulation of flow rates, such as fetal urine production and swallowing; and (iii) maternal effects on transplacental fluid movement. Total water accumulation in utero during pregnancy reaches about 4 L (fetus 2,800 mL; placenta 400 mL; AF 800 mL).8 Urine production per kg of body weight increases from 110 mL/kg/24 hour at 25 weeks to 190 mL/kg/24 hour at 35 weeks.17 Interference with disposal in the routes of fluid production by a factor affecting only 1 percent of the volume may increase or decrease total AFV by as much as 1 L in 10 days. AF turnover continues even after fetal death, but it is reduced by about 50 percent,18 implying that membranes may be responsible for about half of the water exchange. This suggests that the membranes play a larger role in water disposal than in production. Indeed, electron microscopic studies19 correlate with an absorptive function of the membranes. It is unlikely that excess AF production results solely from excess urine production or a failure of the fetus to swallow AF.20 The amnion must play a role in the maintenance of AFV and composition. Earlier studies concluded that 25–50 percent of the fluid turnover takes place through the fetus in late pregnancy.21 Abramovich22 challenged the concept that swallowing and voiding are important in controlling the AFV. He showed that some anencephalics may swallow considerable amounts of AF

and that normal volumes were found in esophageal atresia and in the absence of fetal kidneys. Thus, other factors are involved in controlling the AFV. Chamberlain23 has reviewed the studies done on abnormalities of AFV and altered perinatal outcome. Ultrasonic assessment of fetal kidney function in normal and complicated pregnancies revealed that the fetal urinary production rate was 2.2 mL/h at 22 weeks, increasing to 26.3 mL/h at 40 weeks.24 The authors concluded that regulation by the central nervous system does not play a large role in fetal urination control, and that fetal polyuria does not explain polyhydramnios. Polyhydramnios was accompanied by elevated AF pressures.25 Various techniques have been used for the direct estimation of AFV. Comparable results have been reported using dilution techniques, radioactive materials, or various dyes or chemicals.26–33 Abnormal AFV is associated with increased maternal risk and perinatal morbidity and mortality, but the invasive nature of AFV assessment limited its clinical utility.34 The vertical pocket measurement (VPM) is simple but remains semiquantitative with limited accuracy. The AF Index (AFI) is the result of the sum of the four maximum vertical pockets (MVP) from each quadrant of the uterus. A meta-analysis concluded that both AFI and VPM identified abnormal AFVs poorly. The AFI led to more false-positive oligohydramnios findings, and more interventions without improvement in perinatal outcome.35 Population differences for the AFI may also exist.36 Sandlin et al.37 established reference ranges for AFV from 16 to 41 weeks, using dye-dilution techniques and a quantile regression statistical approach (Table 3.1). Refinements in quantifying the noninvasive sonographic assessment of AFV have not significantly improved the predictive ability to identify at-risk pregnancies.38, 39 Polyhydramnios occurs in 1–2 percent of all pregnancies35 and is associated with fetal malformations in about 40 percent of cases.40 Moise defined polyhydramnios in singleton or twin pregnancies as MVP >8 cm in the late second and the third trimesters, and oligohydramnios as MVP 200,000 cells/mL at 16 weeks.5 The number of colony-forming cells is much lower.

CHAPTER 3

Amniotic Fluid Constituents, Cell Culture, and Neural Tube Defects 167

Culture methods The principal difference between the flask and in situ methods resides in the trypsinization step required for a suspension-type harvest. The resulting metaphase spreads can no longer be traced back to an individual parental colony. In contrast, the in situ method leaves colonies intact. When large numbers of cells are required for biochemical or molecular diagnostic studies, culture in T25 flasks remains the method of choice.

10 > 20 CPD 1.5 ± 1.9 n = 20

Frequency

5

3.5 ± 1.8 n = 20

5

0

–1

–2 –3 –4 –5 –6 Clones per mL fluid

–7

–8

Figure 3.9 Cloning efficiency of 20 consecutive amniotic fluid specimens (18 weeks gestational age). Fewer than half of the colonies grew to more than 106 cells per clone (more than 20 cumulative population doublings (CPD)).

Figure 3.9 shows that in platings of 16- to 18-week fluids, an average of 3.5 clones/mL are typically scored at day 12. Only 1.5 colonies per mL reach a clone size of at least 106 cells. Other laboratories report similar values.603 In a series of 14- to 16-week amniocentesis specimens, Hoehn et al.604 observed 3.1 colonies per mL but most were large colonies at day 12. Kennerknecht et al.605 reported high clone counts in 7- to 9-week AF, ranging from 7.9 to 12.2 colonies per mL. Late pregnancy fluids show cloning efficiency of less than 1.5 colonies per mL. Since human AF contains traces of growth and attachment factors such as epidermal growth factor, interleukin-1, tumor necrosis factor 𝛼, fibronectin, and endothelin-1,606 a 1 : 1 mixture of native fluid and growth medium has been recommended.607 Cell growth inhibitors (e.g. IGFBP-1, an insulin-like growth factor binding protein) have also been found in human AF.608 Although the proportion of erythrocytes may vary from 103 to 108 cells/mL, only the most severe blood contamination significantly retards or prevents cell growth. Such specimens can be treated before culture with 0.7 percent sodium citrate hypotonic solution or ammonium chloride lysing reagent (BD Biosciences).

In situ procedure Since the early 1980s, in situ culture and harvest has become the preferred method for cytogenetic studies.609–614 The main advantages are: (i) earlier culture harvest leading to a faster diagnosis, (ii) clonal (or, more correctly, colony) analysis leading to an easier distinction between genuine mosaicism and pseudomosaicism, and (iii) recognition of maternal cell contamination on the basis of clonal morphology. Maternal cell contamination (MCC) occurs in up to 0.5 percent of AFC cultures.615–617 To minimize MCC, some laboratories prefer to discard the first 2 mL of AF. PCR-detectable MCC of AF samples has been described as common (4–17 percent of samples) and probably represents contamination by maternal blood. This contamination can be an important consideration for biochemical or molecular genetic studies.618, 619 However, our local experience is that none of 66 direct AF samples have exhibited variable number of tandem repeat (VNTR)-detectable MCC. This is consistent with the 0.5 percent rate of MCC in AFC cultures identified by PCR analysis by Smith et al.620 This is also consistent with our local experience of finding MCC in 21 of 5,108 (0.41 percent) consecutive AFC karyotype studies (i.e. one or two 46,XX colonies among 15 or more 46,XY colonies in the in situ harvests). MCC rarely confounds the interpretation of cytogenetic results. Guidelines and tables are available detailing the number of metaphases to analyze by either suspension or in situ harvests, to exclude mosaicism at a desired confidence level.603, 621–624 A deficit of these calculations is that they ignore artifactual loss of chromosomes, which is more frequent with suspension than with in situ preparations. Environmental conditions (e.g. relative humidity and temperature) during drying of chromosome

168 Genetic Disorders and the Fetus

spreads can influence chromosome spreading, and artifactual aneuploidy is well documented.625, 626 Spurbeck has video-documented the effects of temperature and humidity on metaphase cell spreading.627 To search for mosaicism, the number of colonies sampled is more informative than the number of metaphases analyzed.628, 629 Whether the gold standard should be a 15-colony analysis has been the subject of some debate.630, 631 Guidelines issued by the American College of Medical Genetics632 recommend for the flask technique counting 20 cells from at least two independently established cultures, analyzing five and karyotyping two. For the in situ method, counting a minimum of 15 cells from at least 15 colonies in at least two independently established cultures was recommended. Laboratories using the in situ technique in conjunction with optimal culture media (e.g. Chang or AmnioMAX from Irvine Scientific or GIBCO, respectively) are able to karyotype most specimens in less than two weeks. Longer time intervals result from suboptimal cell growth conditions, adherence to a 5-day work week, or from other administrative rather than biologic limitations. Many laboratories also employ a robotic harvesting system and an environmental control chamber to improve the number and quality of metaphase cells.625, 633, 634 A typical AFC culture protocol was published by Miron in 2012.633 Automated harvesting for in situ chromosome analysis usually employs a Tecan or Scinomix Sci-Prep robotic sample processor,635 which saves personnel time and improves consistency because the timing, rate, and quantity of aspiration and dispensing of media, hypotonic solution, and fixative are automated. An environmental room or chamber that controls temperature, humidity, and airflow is helpful to both optimize the quality of the metaphase spreading and reduce seasonal variations in harvest quality. The system must be optimized in each laboratory but generally provides high-quality preparations in the range of 35–45 percent relative humidity at 25∘ C.625

examined whether maternal activity or uterine agitation before amniocentesis affect the concentration of viable amniocytes in the fluid.636, 637 After correcting for gestational age, both studies were negative. Enrichment of the cell culture innoculum has been attempted via centrifugation of fluids through isopycnic gradients.638 With bloody specimens, these methods might be of some help but they are impractical for routine use. This limitation also holds for an enrichment technique by which the AF is returned to the fetus after aspiration, filtration, and reinjection of AF as early as 12.5 gestational weeks.639–642

Enhancement of amniotic fluid cell growth

Reduction of oxygen supply Other efforts directed at improving the cellular microenvironment take into account that atmospheric oxygen conditions are not optimal for most mammalian cells in culture. Brackertz et al.646 and Held and Sönnichsen647 demonstrated improved

Enrichment techniques Since cloning efficiency is low, it would be advantageous to increase the number of viable cells introduced into the cell cultures. Two studies have

Growth on extracellular matrix surface The culture surface has a definite influence on the rate of attachment and proliferation. To attach to the culture surface, AFCs must create their own microenvironment, consisting of glycoproteins, collagen, laminin, and fibronectin, among others (ECM proteins). Fetal bovine serum contains fibronectin and so does human AF at 15–18 weeks of gestation.643 If serum concentrations of less than 10 percent are used (such as in Chang-type media supplements), the presence of AF in the culture setup might facilitate the coating of fibronectin on plastic surfaces. Chang and Jones607 reported optimal cloning and growth when cultures were initiated with equal parts of AF and growth factor-supplemented medium (including 4 percent fetal bovine serum). Two other studies have shown that precoating the plastic surfaces with ECM improves both cloning and rate of growth of AFCs.644, 645 In both laboratories, ECM-coated dishes were custom made from bovine corneal endothelial cells. The use of such precoated surfaces may be advantageous for cell attachment and cloning if suboptimal media have to be used. It appears impractical for routine use unless the laboratory is prepared to accept the extra expense involved in purchasing precoated dishes. A number of manufacturers offer such “biologic” plastic ware.

CHAPTER 3

Amniotic Fluid Constituents, Cell Culture, and Neural Tube Defects 169

AFC growth under hypoxic conditions. If multigas incubators are opened frequently, however, their humidity tends to decrease, which could disadvantage cells growing in open dishes. Testing and handling fetal bovine serum

The single most important factor in determining the speed and success of prenatal cytogenetic diagnosis is the quality of the growth medium and its supplements.557, 607 Since the traditional medium supplement, fetal bovine serum, represents a complex mixture of growth-promoting substances, considerable effort has been directed toward formulating serum-free media in mammalian cell culture.648 Human AFC culture has benefited greatly from the success of these efforts.649, 650 Fetal bovine serum or Chang-type medium, which includes serum, requires proper storage and handling to preserve its effectiveness. Freeze–thaw cycles and exposure to light are particular problems.651 Defined growth factor supplements

The commercial versions of the growth factorsupplemented media are based on the formulation provided by Chang et al.652, 653 The classic “Chang medium” included transferrin, selenium, insulin, tri-iodothyronine, glucagon, fibroblast growth factor, hydrocortisone, testosterone, estradiol, and progesterone. These factors are added to a 1 : 1 mixture of Dulbecco’s modified Eagles’ medium (DMEM) and Ham’s F12 medium, plus sodium bicarbonate, and small amounts of HEPES buffer and antibiotics. Chang et al. pointed out that their preferred basic medium mixture can be replaced by a number of other formulae (e.g. Ham’s F10 or F12, Coon’s modified Ham’s F12, McCoy’s 5A, RPMI 1640, DMEM, minimal essential medium and TC 199) without detriment. Chang and AmnioMAX media that differ in their buffering systems are available for use in closed or open cell culture systems. As with other aspects of the cell culture art and science, local preferences vary with respect to choice of specialized media, fetal or newborn bovine serum, and whether to mix these media with less costly media.609, 633, 634, 653, 654 It is assumed that the various peptides, hormones, and trace elements act synergistically on the recruitment of cells into the cycle and keep

them from reverting to the G0 stage after completed division. A greater number of cells within a colony will therefore stay in the proliferative pool. The cycle time of individual cells, with the possible exception of the duration of the G1 phase, is not likely to change. Unless Claussen’s micropipette method655 is used, a culture period of 5–7 days will thus remain the minimum time requirement for prenatal cytogenetic diagnosis employing AFC cultures. In our laboratory, we experimented with 12-hour Colcemid exposure and very early harvests. We obtained a small number of metaphase cells after 3 and even 2 days in cell culture but the number of metaphases was insufficient for a complete analysis. A drawback noted by some users of Chang and AmnioMAX media – other than the expense – is their limited shelf-life. Lyophilized or other, more stable media supplements are offered by some manufacturers (e.g. Condimed, UltroSer) but cloning efficiency testing so far has failed to identify a commercial product that consistently yields higher cloning efficiencies than fresh lots of Chang media.604 Use of Chang-type media may augment the incidence of chromosome breakage and chromosomal mosaicism in AFC cultures but rarely to the extent that the cytogenetic interpretation is compromised.656–659 This may in part result from the fact that Chang media can facilitate the growth of E-type colonies and these colonies yield higher rates of random chromosome changes.588, 611 However, the advantages gained by the reduction of turnaround time and the substantial decrease of culture failures using Chang-type media appear to outweigh the potential drawbacks of increased chromosomal breakage and pseudomosaicism. Culture failure

For most laboratories today, reports are completed in 6–14 days with a mean of 7–11 days to the final report, and the rate of culture failure is below 1 percent, depending somewhat on the timing of the amniocentesis,660 and in many laboratories averages closer to 0.1–0.2 percent (van Dyke, unpublished data). There are multiple reasons for cell culture problems and outright failure.661 With the increased experience of the obstetrician and the universal use of high-resolution ultrasound, maternal urine is now rarely received as an AF sample. Anecdotal evidence of some labs suggests

170 Genetic Disorders and the Fetus

that the risk of culture failure is higher in cases of fetal aneuploidy. In one published retrospective study, 56 (0.7 percent) of 7,872 AF samples did not grow.662 Twenty-four of these were judged technically inadequate and 10 were from women whose fetuses had died. Of the remaining 32 cases, 4 had proven (determined by repeat amniocentesis) and 4 had possible (extrapolated from fetal phenotype) aneuploidy. This 25 percent rate of growth failure associated with proven or likely chromosomal aberrations was not confirmed in a similar study comprising 6,369 cases and a growth failure rate of 1.2 percent.663 A study of 14,615 cases identified a higher incidence of culture failure in advanced pregnancies with abnormal ultrasound findings but no association with aneuploidy.664 In addition to a baseline level of less than 1 percent unexplained culture failure (the standard set by the American College of Medical Genetics is 2 percent),623, 624, 665 a number of known hazards can interfere with cell growth.

Syringe toxicity and delayed transportation One serious hazard is transmittal of AF in toxic syringes or tubes.666, 667 AF samples should not be transported in syringes; rather, the fluid should always be promptly transferred and transported in conical centrifuge tubes with plastic caps, spinal tap tubes or similar specimen transport containers. Rubber-capped tubes and stoppered syringes should not be used as storage or transport containers for AF. Problems reported in the United States prompted one manufacturer to recommend minimizing both the time of AF in the syringe and contact with the stopper attached to the plunger rod. Although it is advisable to deliver AF samples to the laboratory without delay, in our experience with AF specimens transported by courier and various delivery services, cell viability is maintained for at least 5 days, assuming the sample is not exposed to extreme temperatures. There is one report of successful cell cultures after unfortunate delays of more than 2 weeks.668 Microbial contamination Microbial contamination is a rare cause of culture failure in experienced laboratories and is largely preventable. As noted earlier, AF itself has bacteriocidal properties. If overwhelming microbial

contamination is apparent within 24 hours after setup, it is probably due to improper handling of the specimen between amniocentesis and delivery to the laboratory (e.g. leakage from loose screw caps or poorly packaged syringes). Approximately 10–20 percent of all samples are cell rich and their turbidity should not be a source of anxiety with regard to possible contamination. This also holds for brownish fluids containing cellular debris and granules in addition to erythrocytes. Seguin and Palmer669 measured cell growth from clear, cloudy (cell-rich), bloody, and dark brown fluids. They showed that cloudy fluids yield better growth than clear ones. They confirmed earlier observations670 that very bloody fluids adversely affect the cloning efficiency. If bacterial or yeast contamination arises during the course of cell culture, it is by no means hopeless to attempt to salvage such cultures. Penicillin-, streptomycin-, or fungicide-supplemented media are used to feed cultures daily after initial frequent washings. Increased chromosomal breakage rates and elevated rates of pseudomosaicism may be observed in such salvaged cultures but if the metaphase cells are analyzable, this rarely interferes with interpretation of the results.

Mycoplasma Mycoplasma is not a significant problem in AFC culture, due mainly to better quality control by the serum manufacturers but also to the awareness of cell culturists that AFC cultures should not share incubator space with established cell lines. A shared water bath used for heating media and trypsin can be a source of mycoplasma contamination because permanent cell lines, frequently shipped from laboratory to laboratory, remain the prevailing source of mycoplasma contamination. As additional protection, many laboratories heat-inactivate their sera before use. Commercial test kits are available for the detection of mycoplasma infections in cell cultures.671 Plastic ware and media storage There have been occasional batches of cell culture-grade plastic that do not, or barely, support cell attachment and growth. As with any component of the cell culture system, it is advisable to test new and old plastic ware in parallel for toxicity and ability to support growth in vitro.

CHAPTER 3

Amniotic Fluid Constituents, Cell Culture, and Neural Tube Defects 171

Hoehn’s laboratory switched several times between Corning and Falcon and tested additional brands because of considerable fluctuations in quality.604

Incubator failure Incubator failure is not a trivial cause of culture loss. The main threats are breakdown of the gas supply or equipment. AF cells cannot tolerate a pH close to or higher than 8 for more than 6–8 hours. On the other hand, a pH of less than 7.0 (for example, due to excess CO2 in the incubator) causes cells to stop dividing. A second danger is overheating of incubators due to mechanical failure or human error. Connection of incubators to emergency power sources is important. Temperature- and gas-sensitive alarm systems are advisable. Record keeping and quality control With the advent of highly standardized cell culture methods, culture hazards have become a much rarer cause for concern in the prenatal diagnosis laboratory. Due to the greater number of specimens processed by the average laboratory, a variety of quality-control measures need to be followed to avoid mistakes ranging from culture mixups to diagnostic errors. The most common and potentially serious laboratory errors are human errors in labeling and cross-contamination of samples. Labeling errors can occur at any stage where cells are transferred between vessels: in the amniocentesis procedure room, at culture initiation, feeding and subculture, harvest, slide making, and even microscope analysis. Cross-contamination of cells between patient samples is most common at the time of cell culture harvest, especially for suspension harvests. For these reasons, quality control and quality assurance programs must include a nonpunitive recording system for all laboratory events. A regular review of those events should seek patterns of error that can be eliminated by continuing education of laboratory staff or (often more effective) process improvement directed at reducing the opportunity for human error. Laboratory directors and supervisors should be familiar with the College of American Pathologists Laboratory General and Cytogenetics checklists and the American College of Medical Genetics Standards and Guidelines.665 Laboratories should also participate in a peer review system such as the CAP proficiency surveys.

Safety in the laboratory

It is the responsibility of the laboratory director and all the laboratory staff to protect the rights, privacy and health of employees, ancillary staff and patients alike. AF specimens and all cultures up to the stage of fixation should be treated as potentially hazardous. Universal precautions are essential. Available resources include the CAP Safety Checklist and excellent reviews of laboratory safety and management.672–674

Mesenchymal stem cells in amniotic fluid

Multipotent mesenchymal stem cells (MSCs) can be obtained from several tissue sources and are of great interest for their potential uses in gene therapy and tissue repair. Those derived from adult bone marrow or other sources apart from AFCs have some drawbacks including their relative rarity and slow rate of proliferation in vitro. In contrast, MSCs derived from AFCs have distinct advantages.675–679 MSCs comprise about 1 percent of the cells in midtrimester AF and likely derive from fibroblastic F-type cells.675 Recent advances in the isolation and culture of MSCs from AF are welcomed because these cells apparently do not form teratomas and are not tumorigenic even after many passages. Amniotic MSCs proliferate well and have stable normal telomeres, cytogenetics, and cell surface markers of pleuripotency, similar to embryonic stem cells. Amniotic MSCs also circumvent ethical objections associated with the use of embryonic stem cells. Although more research is needed, amniotic MSCs appear to have immunogenic characteristics that are favorable for allogenic transplantation.680 They can be coaxed into differentiation along many lineages such as adipogenic, osteogenic, myogenic, endothelial, neurogenic, pancreatic, and hepatic, and including mesodermal, ectodermal, and endodermal lineages.480, 676, 678, 681–683 These cells are likely to find utility in a wide variety of cellular therapies,481, 675 including anticancer combination therapy.684 In a demonstration of whole-tissue engineering for early repair of congenital malformations, heart valves have been fabricated using amniotic MSCs. The engineered heart valves exhibited normal endothelial surfaces and adequate opening and closing behavior.479

172 Genetic Disorders and the Fetus

Under appropriate conditions, amniotic MSCs can form bone.676 The potential exists for amniotic MSCs to be employed for engineering tissues in time to be implanted shortly after birth to repair

malformations of the heart, skin, bladder, or diaphragm.479, 685–687 This approach is unlikely, at least in the short term, to be helpful in the fetal therapy of spina bifida (see Chapters 29 and 30).

References 1. Parkin FM, Lind T, Cheyne GA. Biochemical and cytological changes in liquor amnii with advancing gestation. J Obstet Gynaecol Br Commonw 1969;76:673. 2. Lind T, Hytten FE. Relation of amniotic fluid volume to fetal weight in the first half of pregnancy. Lancet 1970;i:1147. 3. Bourne GL. The anatomy of the human amnion and chorion. Proc R Soc Med 1966;59:1127. 4. Bourne GL. The human amnion and chorion. London: Lloyd-Luke, 1962. 5. Behrman RE, Parer JT, De Lannoy CW, Jr. Placental growth and the formation of amniotic fluid. Nature 1967;214:678. 6. Seeds AEJ. Dynamics of amniotic fluid. In: Natelson S, Scommegna A, Epstein MB, eds. Amniotic fluid. New York: Wiley, 1974:23. 7. Nicolini U, Fisk NM, Talbert DG, et al. Intrauterine manometry: technique and application to fetal pathology. Prenat Diagn 1989;9:243. 8. Fisk NM, Ronderos-Dumit D, Tannirandorn Y, et al. Normal amniotic pressure throughout gestation. Br J Obstet Gynaecol 1992;99:18. 9. Barbera A, Buscaglia M, Ferrazzi E, et al. Intra-amniotic pressure is not affected by amniocentesis between 13 and 18 weeks of gestation. Eur J Obstet Gynecol Reprod Biol 1993;50:185. 10. Hutchinson DL, Hunter CB, Neslen ED, et al. The exchange of water and electrolytes in the mechanism of amniotic fluid formation and the relationship to hydramnios. Surg Gynecol Obstet 1955;100:391. 11. Pritchard JA. Deglutition by normal and anencephalic fetuses. Obstet Gynecol 1965;25:289. 12. Hutchinson DL, Gray MJ, Plentl AA, et al. The role of the fetus in the water exchange of the amniotic fluid of normal and hydramniotic patients. J Clin Invest 1959;38:971. 13. Gulbis B, Jauniaux E, Jurkovic D, et al. Biochemical investigation of fetal renal maturation in early pregnancy. Pediatr Res 1996;39:731. 14. Thomas CR, Lang EK, Lloyd FP. Fetal pyelography – a method for detecting fetal life. A preliminary report. Obstet Gynecol 1963;22:335. 15. Hibbard BM. Polyhydramnios and oligohydramnios. Clin Obstet Gynecol 1962;5:1044.

16. Brace RA. Amniotic fluid volume and its relationship to fetal fluid balance: review of experimental data. Semin Perinatol 1986;10:103. 17. Lotgering FK,Wallenburg HC. Mechanisms of production and clearance of amniotic fluid. Semin Perinatol 1986;10:94. 18. Tervilae L. Transfer of water from maternal blood to amniotic fluid of live and dead fetuses in health and in some pathological conditions of the mother. A study with tritium-labelled water. Ann Chir Gynaecol Fenn 1964;53:1. 19. Bourne GL, Lacy D. Ultra-structure of human amnion and its possible relation to the circulation of amniotic fluid. Nature 1960;186:952. 20. Wintour EM, Shandley L. Effects of fetal fluid balance on amniotic fluid volume. Semin Perinatol 1993;17: 158. 21. Gray MJ, Neslen ED, Plentl AA. Estimation of water transfer from amniotic fluid to fetus. Proc Soc Exp Biol Med 1956;92:463. 22. Abramovich DR. Fetal factors influencing the volume and composition of liquor amnii. J Obstet Gynaecol Br Commonw 1970;77:865. 23. Chamberlain P. Amniotic fluid volume: ultrasound assessment and clinical significance. Semin Perinatol 1985;9:163. 24. Kurjak A, Kirkinen P, Latin V, et al. Ultrasonic assessment of fetal kidney function in normal andcomplicated pregnancies. Am J Obstet Gynecol 1981;141:266. 25. Weiner CP, Grose C. Prenatal diagnosis of congenital cytomegalovirus infection by virus isolation from amniotic fluid. Am J Obstet Gynecol 1990;163:1253. 26. Ostergard DR. The physiology and clinical importance of amniotic fluid. A review. Obstet Gynecol Surv 1970;25:297. 27. Fuchs F. Volume of amniotic fluid at various stages of pregnancy. Clin Obstet Gynecol 1966;9:449. 28. Nelson MM. Amniotic fluid volumes in early pregnancy. J Obstet Gynaecol Br Commonw 1972;79:50. 29. Wagner G, Fuchs F. The volume of amniotic fluid in the first half of human pregnancy. J Obstet Gynaecol Br Emp 1962;69:131. 30. Rhodes P. The volume of liquor amnii in early pregnancy. J Obstet Gynaecol Br Commonw 1966;73:23.

CHAPTER 3

Amniotic Fluid Constituents, Cell Culture, and Neural Tube Defects 173

31. Abramovich DR. The volume of amniotic fluid in

47. Spaggiari E, Heidet L, Grange G, et al. Prognosis and

early pregnancy. J Obstet Gynaecol Br Commonw

outcome of pregnancies exposed to renin-angiotensin system blockers. Prenat Diagn 2012;32:1071.

1968;75:728. 32. Queenan JT, Thompson W, Whitfield CR, et al. Amni-

48. Sherer DM. A review of amniotic fluid dynamics and

otic fluid volumes in normal pregnancies. Am J Obstet

the enigma of isolated oligohydramnios. Am J Perinatol 2002;19:253.

Gynecol 1972;114:34. 33. Moore TR. Clinical evaluation of amniotic fluid vol-

49. Voxman EG, Tran S, Wing DA. Low amniotic fluid index as a predictor of adverse perinatal outcome. J

ume. Semin Perinatol 1993;17:173. 34. Larmon JE, Ross BS. Clinical utility of amniotic fluid volume assessment. Obstet Gynecol Clin North Am

Perinatol 2002;22:282. 50. HellstromWJ, Kogan BA, Jeffrey RB, Jr., et al. The natural history of prenatal hydronephrosis with normal

1998;25:639. 35. Magann EF, Chauhan SP, Doherty DA, et al. A review of idiopathic hydramnios and pregnancy outcomes.

amounts of amniotic fluid. J Urol 1984;132:947. 51. Sutcliffe RG. The nature and origin of the soluble protein in human amniotic fluid. Biol Rev Camb Philos

Obstet Gynecol Surv 2007;62:795. 36. Lu SC, Chang CH, Yu CH, et al. Reappraisal of normal

Soc 1975;50:1.

amniotic fluid index in an Asian population: analysis of

52. Sutcliffe RG, Brock DJ. Immunological studies on the

27,088 records. Taiwan J Obstet Gynecol 2007;46: 260.

nature and origin of the major proteins in amniotic

37. Sandlin AT, Ounpraseuth ST, Spencer HJ, et al.

fluid. J Obstet Gynaecol Br Commonw 1973;80:721.

Amniotic fluid volume in normal singleton pregnan-

53. Gitlin D, Kumate J, Urrusti J, et al. The selectivity of

cies: modeling with quantile regression. Arch Gynecol

the human placenta in the transfer of plasma proteins from mother to fetus. J Clin Invest 1964;43:1938.

Obstet 2014;289:967. 38. Barrilleaux PS, Magann EF, Chauhan SP, et al. Amni-

54. Baardman ME, Erwich JJ, Berger RM, et al. The ori-

otic fluid index as a predictor of adverse perinatal

gin of fetal sterols in second-trimester amniotic fluid:

outcome in the HELLP syndrome. J Reprod Med

endogenous synthesis or maternal–fetal transport? Am J Obstet Gynecol 2012;207:202 e19.

2007;52:293. 39. Morris RK, Meller CH, Tamblyn J, et al. Associa-

55. Muller-Eberhard U, Bashore R. Assessment of

tion and prediction of amniotic fluid measurements

Rh disease by ratios of bilirubin to albumin and

for adverse pregnancy outcome: systematic review and

hemopexin to albumin in amniotic fluid. N Engl J Med 1970;282:1163.

meta-analysis. BJOG 2014;121:686. 40. Stevenson AC, Davison BCC, Oakes MW. Genetic

56. Johnson AM, Umansky I, Alper CA, et al. Amniotic

Lippincott

fluid proteins: maternal and fetal contributions. J Pedi-

41. Moise KJ, Jr. Toward consistent terminology: assess-

57. Heikinheimo M, Jalanko H, Leisti J, et al. Amni-

ment and reporting of amniotic fluid volume. Semin

otic fluid pregnancy-specific beta 1-glycoprotein

Perinatol 2013;37:370.

(SP1) in fetal developmental disorders. Prenat Diagn

counselling,

2nd

edn.

Philadelphia:

Williams & Wilkins, 1976.

42. Beischer N, Desmedt E, Ratten G, et al. The significance of recurrent polyhydramnios. Aust N Z J Obstet Gynecol 1993;33:25. 43. Scott JS. The volume and circulation of the liquor amnii: clinical observations. Proc R Soc Med 1966;59:1128. 44. Esplin MS, Hallam S, Farrington PF, et al. Myotonic dystrophy is a significant cause of idiopathic polyhydramnios. Am J Obstet Gynecol 1998;179:974.

atr 1974;84:588.

1984;4:147. 58. Jorgensen OS, Norgaard-Pedersen B. The synaptic membrane D2-protein in amniotic fluid from pregnancies with fetal neural tube defects. Prenat Diagn 1981;1:3. 59. Sindic CJ, Freund M, Van Regemorter N, et al. S-100 protein in amniotic fluid of anencephalic fetuses. Prenat Diagn 1984;4:297. 60. Anneren G, Esscher T, Larsson L, et al. S-100 protein

45. Benirschke K, McKay DG. The antidiuretic hormone

and neuron-specific enolase in amniotic fluid as mark-

in fetus and infant; histochemical observations with

ers of abdominal wall and neural tube defects in the

special reference to amniotic fluid formation. Obstet Gynecol 1953;1:638. 46. Potter EL. Bilateral absence of ureters and kidneys: a report of 50 cases. Obstet Gynecol 1965;25:3.

fetus. Prenat Diagn 1988;8:323. 61. Gogiel T, Bielecki DA, Bankowski E. Collagenous constituents of amniotic fluid. Acta Biochim Pol 1998;45:1037.

174 Genetic Disorders and the Fetus

62. Uyeno D. The physical properties and chemical composition of human amniotic fluid. J Biol Chem 1919; 37:77. 63. Campbell J, Wathen N, Macintosh M, et al. Biochemical composition of amniotic fluid and extraembryonic coelomic fluid in the first trimester of pregnancy. Br J Obstet Gynaecol 1992;99:563. 64. Chiu RW, Chan KC, Gao Y, et al. Noninvasive prenatal diagnosis of fetal chromosomal aneuploidy by massively parallel genomic sequencing of DNA in maternal plasma. Proc Natl Acad Sci USA 2008;105:20458. 65. Hui L, Wick HC, Edlow AG, et al. Global gene expression analysis of term amniotic fluid cell-free fetal RNA. Obstet Gynecol 2013;121:1248. 66. Salafsky IS, Nadler HL. Intracellular organelles and enzymes in cell-free amniotic fluid. Am J Obstet Gynecol 1971;111:1046. 67. Risteli L, Autio-Harmainen H, von Koskull H, et al. Basement membrane proteins in human amniotic fluid. Clin Genet 1984;26:271. 68. Phimister GM, Marshall RD. Tamm-Horsfall glycoprotein in human amniotic fluid. Clin Chim Acta 1983;128:261. 69. Lee TY, Schafer IA. Glycosaminoglycan composition of human amniotic fluid. Biochim Biophys Acta 1974;354:264. 70. Duncan DM, Logan RW, Ferguson-Smith MA, et al. The measurement of acid mucopolysaccharides (glycosaminoglycans) in amniotic fluid and urine. Clin Chim Acta 1973;45:73. 71. Thompson AR. Factor IX and prothrombin in amniotic fluid and fetal plasma: constraints on prenatal diagnosis of hemophilia B and evidence of proteolysis. Blood 1984;64:867. 72. Queenan JT, Gadow EC, Bachner P, et al. Amniotic fluid proteins in normal and Rh-sensitized pregnancies. Am J Obstet Gynecol 1970;108:406. 73. Jauniaux E,Gulbis B, Hyett J, et al. Biochemical analyses of mesenchymal fluid in early pregnancy. Am J Obstet Gynecol 1998;178:765. 74. Lambotte R, Uhlenbruck G. Amniomucoids – a new class of hexosamine-rich glycoproteins. Nature 1966; 212:290. 75. Lambotte R, Gosselin-Ray C. Analyse électrophorétique de la teneur en polysaccharides de l’𝛼1 globuline spécifique au liquide amniotique humain. Arch Int Physiol Biochim Biophys 1967;75:109. 76. Chitayat D, Marion RW, Squillante L, et al. Detection and enumeration of colonic mucosal cells in amniotic fluid using a colon epithelial-specific monoclonal antibody. Prenat Diagn 1990;10:725.

77. Friel LA, Romero R, Edwin S, et al. The calcium binding protein, S100B, is increased in the amniotic fluid of women with intra-amniotic infection/inflammation and preterm labor with intact or ruptured membranes. J Perinat Med 2007;35:385. 78. Tskitishvili E, Komoto Y, Temma-Asano K, et al. S100B protein expression in the amnion and amniotic fluid in pregnancies complicated by pre-eclampsia. Mol Hum Reprod 2006;12:755. 79. Florio P, Michetti F, Bruschettini M, et al. Amniotic fluid S100B protein in mid-gestation and intrauterine fetal death. Lancet 2004;364:270. 80. Cho CK, Shan SJ, Winsor EJ, et al. Proteomics analysis of human amniotic fluid. Mol Cell Proteomics 2007;6:1406. 81. Michaels JE, Dasari S, Pereira L, et al. Comprehensive proteomic analysis of the human amniotic fluid proteome: gestational age-dependent changes. J Proteome Res 2007;6:1277. 82. Buhimschi CS, Buhimschi IA, Abdel-Razeq S, et al. Proteomic biomarkers of intra-amniotic inflammation: relationship with funisitis and early-onset sepsis in the premature neonate. Pediatr Res 2007;61:318. 83. Buhimschi IA, Zhao G, Rosenberg VA, et al. Multidimensional proteomics analysis of amniotic fluid to provide insight into the mechanisms of idiopathic preterm birth. PLoS One 2008;3:e2049. 84. Kamath-Rayne BD, Smith HC, Muglia LJ, et al. Amniotic fluid: the use of high-dimensional biology to understand fetal well-being. Reprod Sci 2014;21:6. 85. Wang TH, Chang YL, Peng HH, et al. Rapid detection of fetal aneuploidy using proteomics approaches on amniotic fluid supernatant. Prenat Diagn 2005;25:559. 86. Hassan MI, Kumar V, Singh TP, et al. Proteomic analysis of human amniotic fluid from Rh(−) pregnancy. Prenat Diagn 2008;28:102. 87. Biezenski JJ. Incorporation of 14 C-1-palmitate into rabbit fetal lipids in vivo. Am J Obstet Gynecol 1976;126:356. 88. Robertson AF, Sprecher H. A review of human placental lipid metabolism and transport. Acta Paediatr Scand 1968:Suppl 183:3. 89. Pomerance W, Biezenski JJ, Moltz A, et al. Origin of amniotic fluid lipids. II. Abnormal pregnancy. Obstet Gynecol 1971;38:379. 90. Gardella C, Hitti J, Martin TR, et al. Amniotic fluid lipopolysaccharide-binding protein and soluble CD14 as mediators of the inflammatory response in preterm labor. Am J Obstet Gynecol 2001;184:1241. 91. Dallaire L, Mitchell G, Giguere R, et al. Prenatal diagnosis of Smith-Lemli-Opitz syndrome is possible

CHAPTER 3

Amniotic Fluid Constituents, Cell Culture, and Neural Tube Defects 175

by measurement of 7-dehydrocholesterol in amniotic fluid. Prenat Diagn 1995;15:855. 92. Tint GS, Abuelo D, Till M, et al. Fetal Smith-LemliOpitz syndrome can be detected accurately and reliably by measuring amniotic fluid dehydrocholesterols. Prenat Diagn 1998;18:651. 93. Wassif CA, Maslen C, Kachilele-Linjewile S, et al. Mutations in the human sterol delta7-reductase gene at 11q12-13 cause Smith-Lemli-Opitz syndrome. Am J Hum Genet 1998;63:55. 94. Loeffler J, Utermann G, Witsch-Baumgartner M. Molecular prenatal diagnosis of Smith-Lemli-Opitz syndrome is reliable and efficient. Prenat Diagn 2002;22:827. 95. Waye JS, Eng B, Nowaczyk MJ. Prenatal diagnosis of Smith-Lemli-Opitz syndrome (SLOS) by DHCR7 mutation analysis. Prenat Diagn 2007;27:638. 96. Shinawi M, Szabo S, Popek E, et al. Recognition of Smith-Lemli-Opitz syndrome (RSH) in the fetus: utility of ultrasonography and biochemical analysis in pregnancies with low maternal serum estriol. Am J Med Genet A 2005;138:56. 97. Chevy F, Humbert L, Wolf C. Sterol profiling of amniotic fluid: a routine method for the detection of distal cholesterol synthesis deficit. Prenat Diagn 2005;25:1000. 98. Fitzky BU, Witsch-Baumgartner M, Erdel M, et al. Mutations in the Delta7-sterol reductase gene in patients with the Smith-Lemli-Opitz syndrome. Proc Natl Acad Sci U S A 1998;95:8181. 99. Yu H, Lee MH, Starck L, et al. Spectrum of delta(7)-dehydrocholesterol

reductase

mutations

in patients with the Smith-Lemli-Opitz (RSH) syndrome. Hum Mol Genet 2000;9:1385. 100. Yu H, Tint GS, Salen G, et al. Detection of a common mutation in the RSH or Smith-Lemli-Opitz syndrome by a PCR-RFLP assay: IVS8-G→C is found in over sixty percent of US propositi. Am J Med Genet 2000;90:347. 101. Haas D, Haege G, Hoffmann GF, et al. Prenatal presentation and diagnostic evaluation of suspected Smith-Lemli-Opitz (RSH) syndrome. Am J Med Genet A 2013;161A:1008. 102. Goldenberg A, Wolf C, Chevy F, et al. Antenatal manifestations of Smith-Lemli-Opitz (RSH) syndrome: a retrospective survey of 30 cases. Am J Med Genet A 2004;124A:423. 103. Hagenfeldt L, Hagenfeldt K. Individual free fatty acids in amniotic fluid and in plasma of pregnant women. Br J Obstet Gynaecol 1976;83:383.

104. Rueda R, Vargas ML, Garcia-Pacheco M, et al. Detection of immunoregulatory lipid-like factors in human amniotic fluid. Am J Reprod Immunol 1990;24:40. 105. Heikkinen J, Maentausta O, Tuimala R, et al. Amniotic fluid bile acids in normal and pathologic pregnancy. Obstet Gynecol 1980;56:60. 106. Deleze G, Sidiropoulos D, Paumgartner G. Determination of bile acid concentration in human amniotic fluid for prenatal diagnosis of intestinal obstruction. Pediatrics 1977;59:647. 107. Shrand H. Vomiting in utero with intestinal atresia. Pediatrics 1972;49:767. 108. Gluck L, Kulovich MV. Lecithin-sphingomyelin ratios in amniotic fluid in normal and abnormal pregnancy. Am J Obstet Gynecol 1973;115:539. 109. Gluck L. Surfactant: 1972. Pediatr Clin North Am 1972;19:325. 110. Briand RL, Harold S, Blass KG. High-performance liquid chromatographic determination of the lecithin/sphingomyelin ratio in amniotic fluid. J Chromatogr 1981;223:277. 111. Ghidini A, Poggi SH, Spong CY, et al. Role of lamellar body count for the prediction of neonatal respiratory distress syndrome in non-diabetic pregnant women. Arch Gynecol Obstet 2005;271:325. 112. Karcher R, Sykes E, Batton D, et al. Gestational age specific predicted risk of neonatal respiratory distress syndrome using lamellar body count and surfactant-to-albumin ratio in amniotic fluid. Am J Obstet Gynecol 2005;193:1680. 113. ACOG Committee opinion no. 560: Medically indicated late-preterm and early-term deliveries. Obstet Gynecol 2013;121:908. 114. Damron DP. Fetal lung maturity testing. Am J Obstet Gynecol 2014;211:184. 115. Southren AL, Kobayashi Y, Brenner P, et al. Diamine oxidase activity in human maternal and fetal plasma and tissues at parturition. J Appl Physiol 1965;20:1048. 116. Lapan B, Friedman MM. Enzymes in the amniotic fluid and maternal serum. A comparative study. Am J Obstet Gynecol 1962;83:1337. 117. Usategui-Gomez M. Immunoglobulins and other proteins in amniotic fluid. In: Natelson S, Scommegna A, Epstein MB, eds. Amniotic fluid. New York: Wiley, 1974:111. 118. Uuspaa VJ. High histaminase activity of human blood in pregnancy and the so-called placenta haemochorialis: preliminary report. Ann Med Exp Biol Fenn 1951;29:81. 119. Kerenyi T, Sarkozi L. Diagnosis of fetal death in utero by elevated amniotic fluid CPK levels. Obstet Gynecol 1974;44:215.

176 Genetic Disorders and the Fetus

120. Geyer H, Schneider I. Enzyme in Fruchtwasser [Enzymes of amniotic fluid]. Z Klin Chem Klin Biochem 1970;8:141. 121. Geyer H. Zie herkunft der Fruchtwasser-enzyme [The origin of amniotic fluid enzymes]. Z Klin Chem Klin Biochem 1970;8:145. 122. Potier M, Dallaire L, Melancon SB. Amniotic fluid alpha-galactosidase activity: an indicator of gestational age. Gynecol Invest 1974;5:306. 123. Bacigalupo G, Meraner R. Pyruvate kinase in human amniotic fluid – a new indicator of fetal maturity in late pregnancy. J Perinat Med 1984;12:97. 124. Brocklehurst D, Wilde CE. Amniotic fluid alkaline phosphatase, gamma-glutamyltransferase, and 5′ -nucleotidase activity from 13 to 40 weeks’ gestation, and alkaline phosphatase as an index of fetal lung maturity. Clin Chem 1980;26:588. 125. Gurdol F, Genc S, Yalcin O, et al. The presence of prolidase activity in amniotic fluid and its evaluation as a maturity test. Biol Neonate 1995;67:34. 126. Butterworth J, Broadhead DM, Sutherland GR, et al. Lysosomal enzymes of amniotic fluid in relation to gestational age. Am J Obstet Gynecol 1974;119:821. 127. Parmley TH, Seeds AE. Fetal skin permeability to isotopic water (THO) in early pregnancy. Am J Obstet Gynecol 1970;108:128. 128. Lind T, Kendall A, Hytten FE. The role of the fetus in the formation of amniotic fluid. J Obstet Gynaecol Br Commonw 1972;79:289. 129. Fluharty AL, Scott ML, Porter MT, et al. Acid alpha-glucosidase in amniotic fluid. Biochem Med 1973;7:39. 130. Potier M, Dallaire L, Melancon SB. Occurrence and properties of fetal intestinal glycosidases (disaccharidases) in human amniotic fluid. Biol Neonate 1975;27:141. 131. Antonowicz I, Milunsky A, Lebenthal E, et al. Disaccharidase and lysosomal enzyme activities in amniotic fluid, intestinal mucosa and meconium. Correlation between morphology and disaccharidase activities in human fetal small intestine. Biol Neonate 1977;32: 280. 132. Nadler HL, Messina AM. In-utero detection of type- II glycogenosis (Pompe’s disease). Lancet 1969;ii: 1277. 133. Nadler HL, Bigley RH, Hug G. Prenatal detection of Pompe’s disease. Lancet 1970;ii:369. 134. Sutcliffe RG, Brock DJ, Robertson JG, et al. Enzymes in amniotic fluid: a study of specific activity patterns during pregnancy. J Obstet Gynaecol Br Commonw 1972;79:895. 135. Butterworth J, Sutherland GR, Bain AD, et al. Lysosomal enzymes in amniotic fluid. Clin Chim Acta 1972;39:275.

136. Lowden JA, Cutz E, Conen PE, et al. Prenatal diagnosis of G M1-gangliosidosis. N Engl J Med 1973;288:225. 137. Gomez-Ospina N. Arylsulfatase A deficiency. In: Adam MP, Ardinger HH, Pagon RA, et al., eds. GeneReviews. http://www.ncbi.nlm.nih.gov/pubmed/ 20301309 (accessed July 7, 2020). 138. Huijing F, Warren RJ, McLeod AG. Elevated activity of lysosomal enzymes in amniotic fluid of a fetus with mucolipidosis II (I-cell disease). Clin Chim Acta 1973;44:453. 139. Poenaru L, Mezard C, Akli S, et al. Prenatal diagnosis of mucolipidosis type II on first-trimester amniotic fluid. Prenat Diagn 1990;10:231. 140. Friedland J, Perle G, Saifer A, et al. Screening for Tay-Sachs disease in utero using amniotic fluid. Proc Soc Exp Biol Med 1971;136:1297. 141. O’Brien JS, Okada S, Fillerup DL, et al. Tay-Sachs disease: prenatal diagnosis. Science 1971;172:61. 142. Desnick RJ, Krivit W, Sharp HL. In utero diagnosis of Sandhoff’s disease. Biochem Biophys Res Commun 1973;51:20. 143. Potier M, Boire G, Dallaire L, et al. N-Acetyl-betahexosaminidase isoenzymes of amniotic fluid and maternal serum. Their relevance to prenatal diagnosis of the GM2 gangliosidoses. Clin Chim Acta 1977;76:309. 144. Guetta E, Peleg L. Rapid detection of fetal Mendelian disorders: Tay-Sachs disease. Methods Mol Biol 2008;444:147. 145. Dallaire L, Gagnon M, Kinch RA. Fetal amino acid metabolism. Clin Res 1971;19:766. 146. Jauniaux E, Gulbis B, Gerloo E. Free amino acids in human fetal liver and fluids at 12–17 weeks of gestation. Hum Reprod 1999;14:1638. 147. Wenstrom KD, Johanning GL, Owen J, et al. Role of amniotic fluid homocysteine level and of fetal 5,10-methylenetetrahydrafolate reductase genotype in the etiology of neural tube defects. Am J Med Genet 2000;90:12. 148. Jauniaux E, Sherwood RA, Jurkovic D, et al. Amino acid concentrations in human embryological fluids. Hum Reprod 1994;9:1175. 149. Jauniaux E, Gulbis B, Gerlo E, et al. Free amino acid distribution inside the first trimester human gestational sac. Early Hum Dev 1998;51:159. 150. Mesavage WC, Suchy SF, Weiner DL, et al. Amino acids in amniotic fluid in the second trimester of gestation. Pediatr Res 1985;19:1021. 151. Dallaire L, Potier M, Melancon SB, et al. Feto-maternal amino acid metabolism. J Obstet Gynaecol Br Commonw 1974;81:761.

CHAPTER 3

Amniotic Fluid Constituents, Cell Culture, and Neural Tube Defects 177

152. Kamoun P, Parvy P, Pham Dinh D, et al. Citrulline in amniotic fluid and the prenatal diagnosis of citrullinemia. Prenat Diagn 1983;3:53. 153. Chadefaux-Vekemans B, Rabier D, Chabli A, et al. Improving the prenatal diagnosis of citrullinemia using citrulline/ornithine+arginine ratio in amniotic fluid. Prenat Diagn 2002;22:456. 154. Kleijer WJ, Blom W, Huijmans JG, et al. Prenatal diagnosis of citrullinemia: elevated levels of citrulline in the amniotic fluid in the three affected pregnancies. Prenat Diagn 1984;4:113. 155. Kleijer WJ, Garritsen VH, van der Sterre ML, et al. Prenatal diagnosis of citrullinemia and argininosuccinic aciduria: evidence for a transmission ratio distortion in citrullinemia. Prenat Diagn 2006;26:242. 156. Northrup H, Beaudet AL, O’Brien WE. Prenatal diagnosis of citrullinaemia: review of a 10-year experience including recent use of DNA analysis. Prenat Diagn 1990;10:771. 157. Pettit BR, King GS, Blau K. The analysis of hexitols in biological fluid by selected ion monitoring. Biomed Mass Spectrom 1980;7:309. 158. Elsas LJ, Lai K, Saunders CJ, et al. Functional analysis of the human galactose-1-phosphate uridyltransferase promoter in Duarte and LA variant galactosemia. Mol Genet Metab 2001;72:297. 159. Coude M, Chadefaux B, Rabier D, et al. Early amniocentesis and amniotic fluid organic acid levels in the prenatal diagnosis of organic acidemias. Clin Chim Acta 1990;187:329. 160. Jakobs C, Ten Brink HJ, Stellaard F. Prenatal diagnosis of inherited metabolic disorders by quantitation of characteristic metabolites in amniotic fluid: facts and future. Prenat Diagn 1990;10:265. 161. Lindblad B, Lindstedt S, Steen G. On the enzymic defects in hereditary tyrosinemia. Proc Natl Acad Sci U S A 1977;74:4641. 162. Gagne R, Lescault A, Grenier A, et al. Prenatal diagnosis of hereditary tyrosinaemia: measurement of succinylacetone in amniotic fluid. Prenat Diagn 1982;2:185. 163. Poudrier J, Lettre F, St-Louis M, et al. Genotyping of a case of tyrosinaemia type I with normal level of succinylacetone in amniotic fluid. Prenat Diagn 1999;19:61. 164. Maydan G, Andresen BS, Madsen PP, et al. TAT gene mutation analysis in three Palestinian kindreds with oculocutaneous tyrosinaemia type II; characterization of a silent exonic transversion that causes complete missplicing by exon 11 skipping. J Inherit Metab Dis 2006;29:620.

165. Antonowicz I, Chang SK, Grand RJ. Development and distribution of lysosomal enzymes and disaccharidases in human fetal intestine. Gastroenterology 1974;67:51. 166. Morin PR, Potier M, Dallaire L, et al. Prenatal detection of the autosomal recessive type of polycystic kidney disease by trehalase assay in amniotic fluid. Prenat Diagn 1981;1:75. 167. Morin PR, Potier M, Dallaire L, et al. Prenatal detection of intestinal obstruction: deficient amniotic fluid disaccharidases in affected fetuses. Clin Genet 1980;18:217. 168. Claass AH, van Diggelen OP, Hauri HP, et al. Characteristics of maltase activity in amniotic fluid. Clin Chim Acta 1985;145:275. 169. Potier M, Morin PR, Melancon SB, et al. Differential stabilities of fetal intestinal disaccharidases determine their relative amounts released into amniotic fluid. Biol Neonate 1984;45:257. 170. Potier M, Cousineau J, Michaud L, et al. Fetal intestinal microvilli in human amniotic fluid. Prenat Diagn 1986;6:429. 171. Jalanko H, Rapola J, Lehtonen E. Particulate fraction in amniotic fluid at second trimester. J Clin Pathol 1985;38:1065. 172. Morin PR, Melancon SB, Dallaire L, et al. Prenatal detection of intestinal obstructions, aneuploidy syndromes, and cystic fibrosis by microvillar enzyme assays (disaccharidases, alkaline phosphatase, and glutamyltransferase) in amniotic fluid. Am J Med Genet 1987;26:405. 173. Morin PR, Potier M, Dallaire L, et al. Prenatal detection of the congenital nephrotic syndrome (Finnish type) by trehalase assay in amniotic fluid. Prenat Diagn 1984;4:257. 174. Poenaru L, Vinet MC, Dreyfus JC. Human amniotic fluid alpha-glucosidase. Clin Chim Acta 1981;117:53. 175. Elsliger MA, Dallaire L, Potier M. Fetal intestinal and renal origins of trehalase activity in human amniotic fluid. Clin Chim Acta 1993;216:91. 176. Dallaire L, Perreault G. Hereditary multiple intestinal atresia. Birth Defects Orig Artic Ser 1974;10:259. 177. Garel C, Dreux S, Philippe-Chomette P, et al. Contribution of fetal magnetic resonance imaging and amniotic fluid digestive enzyme assays to the evaluation of gastrointestinal tract abnormalities. Ultrasound Obstet Gynecol 2006;28:282. 178. Muller F, Dreux S, Vaast P, et al. Prenatal diagnosis of megacystis-microcolon-intestinal hypoperistalsis syndrome: contribution of amniotic fluid digestive enzyme assay and fetal urinalysis. Prenat Diagn 2005;25:203. 179. Machado L, Matias A, Rodrigues M, et al. Fetal megacystis as a prenatal challenge:

178 Genetic Disorders and the Fetus

180.

181.

182.

183.

184.

185.

186.

187.

188.

189.

190.

191.

192.

193.

194.

megacystis-microcolonintestinal hypoperistalsis syndrome in a male fetus. Ultrasound Obstet Gynecol 2013;41:345. Talabani H, Dreux S, Luton D, et al. Fetal anal incontinence evaluated by amniotic fluid digestive enzyme assay in myelomeningocele spina bifida. Pediatr Res 2005;58:766. Potier M, Melancon SB, Dallaire L. Fetal intestinal disaccharidases in human amniotic fluid. Biomedicine 1976;25:167. Arey LB. Developmental anatomy: a textbook and laboratory manual of embryology, 7th edn. Philadelphia: WB Saunders, 1965. Gitlin D, Kumate J, Morales C, et al. The turnover of amniotic fluid protein in the human conceptus. Am J Obstet Gynecol 1972;113:632. Mulivor RA, Mennuti MT, Harris H. Origin of the alkaline phosphatases in amniotic fluid. Am J Obstet Gynecol 1979;135:77. Pocknee RC, Abramovich DR. Origin and levels of trypsin in amniotic fluid throughout pregnancy. Br J Obstet Gynaecol 1982;89:142. van Diggelen OP, Janse HC, Kleijer WJ. Disaccharidases in amniotic fluid as possible prenatal marker for cystic fibrosis. Lancet 1983;i:817. Jarmas AL, Weaver DD, Padilla LM, et al. Hirschsprung disease: etiologic implications of unsuccessful prenatal diagnosis. Am J Med Genet 1983;16:163. Carbarns NJ, Gosden C, Brock DJ. Microvillar peptidase activity in amniotic fluid: possible use in the prenatal diagnosis of cystic fibrosis. Lancet 1983;i:329. Brock DJ. Amniotic fluid alkaline phosphatase isoenzymes in early prenatal diagnosis of cystic fibrosis. Lancet 1983;ii:941. Szabo M, Munnich A, Teichmann F, et al. Discriminant analysis for assessing the value of amniotic fluid microvillar enzymes in the prenatal diagnosis of cystic fibrosis. Prenat Diagn 1990;10:761. Morin PR, Potier M, LaSalle R, et al. Amniotic-fluid disaccharidases in the prenatal detection of cystic fibrosis. Lancet 1983;ii:621. Janlanko H, Aula P. Decrease in gamma-glutamyl transpeptidase activity in early amniotic fluid in fetal trisomy 18 syndrome. Br Med J 1982;284:1593. Jalanko H. Developmental changes in gamma-glutamyl transpeptidase in human amniotic fluid. Oncodev Biol Med 1983;4:253. Moniz C, Nicolaides KH, Keys D, et al. Gammaglutamyl transferase activity in fetal serum, maternal serum, and amniotic fluid during gestation. J Clin Pathol 1984;37:700.

195. Brock DJ, Bedgood D, Hayward C, et al. Amniotic fluid microvillar enzyme activities in the early detection of fetal abnormalities. Prenat Diagn 1984;4:261. 196. Davidson GP, Cutz E, Hamilton JR, et al. Familial enteropathy: a syndrome of protracted diarrhea from birth, failure to thrive, and hypoplastic villus atrophy. Gastroenterology 1978;75:783. 197. Abramsky O, Brenner T, Lisak RP, et al. Significance in neonatal myasthenia gravis of inhibitory effect of amniotic fluid on binding of antibodies to acetylcholine receptor. Lancet 1979;ii:1333. 198. Brown CL, Colden KA, Hume RF, et al. Faint and positive amniotic fluid acetylcholinesterase with a normal sonogram. Am J Obstet Gynecol 1996;175:1000. 199. Symonds EM, Williams SS, Cellier KM. Maternal and fetal influences on the acid-base balance of human amniotic fluid. Obstet Gynecol 1971;37:742. 200. Jauniaux E, Watson A, Burton G. Evaluation of respiratory gases and acid-base gradients in human fetal fluids and uteroplacental tissue between 7 and 16 weeks’ gestation. Am J Obstet Gynecol 2001;184:998. 201. Vrachnis N, Malamitsi-Puchner A, Samoli E, et al. Elevated mid-trimester amniotic fluid ADAM-8 concentrations as a potential risk factor for preterm delivery. J Soc Gynecol Investig 2006;13:186. 202. Yamashiro C, Kanenishi K, Akiyama M, et al. Adrenomedullin concentrations in early 2nd-trimester amniotic fluid: relation to preterm delivery and fetal growth at birth. Gynecol Obstet Invest 2002;54:99. 203. Gulbis B, Jauniaux E, Cotton F, et al. Protein and enzyme patterns in the fluid cavities of the first trimester gestational sac: relevance to the absorptive role of secondary yolk sac. Mol Hum Reprod 1998;4:857. 204. Jalanko H, Heikinheimo M, Ryynanen M, et al. Alkaline phosphatase activity in amniotic fluid in pregnancies with fetal disorders. Prenat Diagn 1983;3: 303. 205. Evans HE, Glass L, Mandl I. Alpha1-antitrypsin concentration in amniotic fluid. Biol Neonate 1975;27:232. 206. Kleinbauer D, Klink F, Wagner T, et al. Blutgerinnung und Fibrinolyse Faktor-Aktivitäten in der Amnionflüssigkeit. [Blood coagulation and fibrinolytic factor activities in the amniotic fluid]. Geburtshilfe Frauenheilkd 1988;48:397. 207. Chadefaux B, Ceballos I, Rabier D, et al. Prenatal diagnosis of argininosuccinic aciduria by assay of argininosuccinate in amniotic fluid at the 12th week of gestation. Am J Med Genet 1990;35:594. 208. Fernandez de Castro A, Usategui-Gomez M, Spellacy WN. Amniotic fluid amylase. Am J Obstet Gynecol 1973;116:931.

CHAPTER 3

Amniotic Fluid Constituents, Cell Culture, and Neural Tube Defects 179

209. Dawood MY. Hormones in amniotic fluid. Am J Obstet Gynecol 1977;128:576. 210. Madazli R, Atis A, Uzun H, et al. Mid-trimester amniotic fluid angiogenin, lactate dehydrogenase and fibronectin in the prediction of preterm delivery. Eur J Obstet Gynecol Reprod Biol 2003;106:160. 211. Cohen SB, Goldenberg M, Rabinovici J, et al. Anticardiolipin antibodies in fetal blood and amniotic fluid derived from patients with the anti-phospholipid syndrome. Hum Reprod 2000;15:1170. 212. Pacsa AS. Impairment of immunity during pregnancy and antiviral effect of amniotic fluid. Lancet 1977;1:330. 213. Fainaru M, Deckelbaum R, Golbus MS. Apolipoproteins in human fetal blood and amniotic fluid in midtrimester pregnancy. Prenat Diagn 1981;1:125. 214. Ruelland A, Mention JE, Perrot Y, et al. Evolution du taux de l’apolipoproteine A dans le liquide amniotique au cours de la grossesse normale et pathologique [The levels of apolipoprotein A in liquor in normal and pathological pregnancies (author’s transl.)]. J Gynecol Obstet Biol Reprod (Paris) 1982;11:241. 215. Chadefaux-Vekemans B, Rabier D, Cadoudal N, et al. Prenatal diagnosis of some metabolic diseases using early amniotic fluid samples: report of a 15 years, experience. Prenat Diagn 2006;26:814. 216. Borresen AL, van der Hagen CB. Metachromatic leukodystrophy. II. Direct determination of arylsulphatase A activity in amniotic fluid. Clin Genet 1973;4:442. 217. Di Lieto A, Pollio F, Catalano D, et al. Atrial natriuretic factor in amniotic fluid and in maternal venous blood of pregnancies with fetal cardiac malformations and chromosomal abnormalities. J Matern Fetal Neonatal Med 2002;11:183. 218. Lewis JF, Johnson P, Miller P. Evaluation of amniotic fluid for aerobic and anaerobic bacteria. Am J Clin Pathol 1976;65:58. 219. Prevedourakis C, Koumentakou E, Zolotas J, et al. E. coli growth inhibition by amniotic fluid. Acta Obstet Gynecol Scand 1976;55:245. 220. Ford LC, DeLange RJ, Lebherz TB. Identification of a bactericidal factor (B-lysin) in amniotic fluid at 14 and 40 weeks’ gestation. Am J Obstet Gynecol 1977; 127:788. 221. Kofinas GD, Kofinas AD, Pyrgerou M, et al. Amniotic fluid beta-endorphin levels and labor. Obstet Gynecol 1987;69:945. 222. Smith AL, Scanlon J. Amnionic fluid d(2)betahydroxybutyrate and the dysmature newborn infant. Am J Obstet Gynecol 1973;115:569.

223. Gulbis B, Gervy C, Jauniaux E. Amniotic fluid biochemistry in second-trimester trisomic pregnancies: relationships to fetal organ maturation and dysfunction. Early Hum Dev 1998;52:211. 224. Perlman M. Golden liquor amnii. Lancet 1973;i:556. 225. Harper P, Bias WB, Hutchinson JR, et al. ABH secretor status of the fetus: a genetic marker identifiable by amniocentesis. J Med Genet 1971;8:438. 226. Arcilla MB, Sturgeon P. Lewis and ABH substances in amniotic fluid obtained by amniocentesis. Pediatr Res 1972;6:853. 227. Jones MI, Spragg SP, Webb T. Detection of proteins in human amniotic fluid using two-dimensional gel electrophoresis. Biol Neonate 1981;39:171. 228. Dawson EB, Evans DR, Nosovitch J. Third-trimester amniotic fluid metal levels associated with preeclampsia. Arch Environ Health 1999;54:412. 229. Auger P, Marquis G, Dallaire L, et al. Stunted growth Candida albicans in human amniotic fluid in vitro. J Lab Clin Med 1980;95:272. 230. Bussière L, Dumont J, Hubert J. Direct determination of calcium, copper, iron, magnesium, manganese and zinc in amniotic fluid samples using inductively coupled plasma-atomic emission spectrometry. Anal Chim Acta 1989;224:73. 231. Buhimschi IA, Zambrano E, Pettker CM, et al. Using proteomic analysis of the human amniotic fluid to identify histologic chorioamnionitis. Obstet Gynecol 2008;111:403. 232. Tayyar M, Tutus A. The effect of maternal age, parity, and fetal sex on the amniotic fluid and maternal serum levels of CA125, CA19.9, CA15.3, and CEA. Int J Fertil Womens Med 1999;44:256. 233. Hahn P, Skala JP, Seccombe DW, et al. Carnitine content of blood and amniotic fluid. Pediatr Res 1977;11:878. 234. Divers WA, Jr., Wilkes MM, Babaknia A, et al. An increase in catecholamines and metabolites in the amniotic fluid compartment from middle to late gestation. Am J Obstet Gynecol 1981;139:483. 235. Holst RM, Laurini R, Jacobsson B, et al. Expression of cytokines and chemokines in cervical and amniotic fluid: relationship to histological chorioamnionitis. J Matern Fetal Neonatal Med 2007;20:885. 236. Anteby SO, Zukerman H, Gedelia I, et al. Citric acid in amniotic fluid. J Obstet Gynaecol Br Commonw 1973;80:27. 237. Tarim E, Bagis T, Kilicdag EB, et al. Are amniotic fluid C-reactive protein and glucose levels, and white blood cell counts at the time of genetic amniocentesis related with preterm delivery? J Perinat Med 2005;33:524.

180 Genetic Disorders and the Fetus

238. Margarit L, Griffiths AN, Tsapanos V, et al. Amniotic fluid endothelin levels and the incidence of premature rupture of membranes. Int J Gynaecol Obstet 2006;93:18. 239. Knepper MA, Pisitkun T. Exosomes in urine: who would have thought? Kidney Int 2007;72:1043. 240. Cassady G, Hinkley C, Bailey P, et al. Amniotic fluid creatinine in pregnancies complicated by diabetes. Am J Obstet Gynecol 1975;122:13. 241. Mussap M, Fanos V, Pizzini C, et al. Predictive value of amniotic fluid cystatin C levels for the early identification of fetuses with obstructive uropathies. BJOG 2002;109:778. 242. Kristensen K, Larsson I, Hansson SR. Increased cystatin C expression in the pre-eclamptic placenta. Mol Hum Reprod 2007;13:189. 243. Oboh AE, Orsi NM, Campbell J. Amniotic fluid cytokine profile in association with fetal hyperechogenic bowel. Eur J Obstet Gynecol Reprod Biol 2006;128:86. 244. Davis LE, Tweed GV, Chin TD, et al. Interuterine diagnosis of cytomegalovirus infection: viral recovery from amniocentesis fluid. Am J Obstet Gynecol 1971;109:1217. 245. Halperin R, Halpern D, Hadas E, et al. Intrauterine levels of human decidua-associated protein (hDP) 200 in normal pregnancy and missed abortion. Gynecol Obstet Invest 1998;46:150. 246. Espinoza J, Chaiworapongsa T, Romero R, et al. Antimicrobial peptides in amniotic fluid: defensins, calprotectin and bacterial/permeability-increasing protein in patients with microbial invasion of the amniotic cavity, intra-amniotic inflammation, preterm labor and premature rupture of membranes. J Matern Fetal Neonatal Med 2003;13:2. 247. Firgaira FA, Cotton RG, Danks DM, et al. Prenatal determination of dihydropteridine reductase in a normal fetus at risk for malignant hyperphenylalaninemia. Prenat Diagn 1983;3:7. 248. Adama van Scheltema PN, In’t Anker PS, Vereecken A, et al. Biochemical composition of amniotic fluid in pregnancies complicated with twin–twin transfusion syndrome. Fetal Diagn Ther 2005;20:186. 249. Keller S, Rupp C, Stoeck A, et al. CD24 is a marker of exosomes secreted into urine and amniotic fluid. Kidney Int 2007;72:1095. 250. Sciarra JJ, Tagatz GE, Notation AD, et al. Estriol and estetrol in amniotic fluid. Am J Obstet Gynecol 1974;118:626. 251. Ramsey PS, Andrews WW, Goldenberg RL, et al. Elevated amniotic fluid ferritin levels are associated with inflammation-related pregnancy loss following

252.

253.

254.

255.

256.

257.

258.

259.

260. 261.

262.

263.

264.

mid-trimester amniocentesis. J Matern Fetal Neonatal Med 2002;11:302. Sakura M, Nakabayashi M, Takeda Y, et al. Elevated fetal fibronectin in midtrimester amniotic fluid is involved with the onset of preeclampsia. J Obstet Gynaecol Res 1998;24:73. Yoneda S, Sakai M, Sasaki Y, et al. Interleukin-8 and glucose in amniotic fluid, fetal fibronectin in vaginal secretions and preterm labor index based on clinical variables are optimal predictive markers for preterm delivery in patients with intact membranes. J Obstet Gynaecol Res 2007;33:38. Clements JA, Reyes FI, Winter JS, et al. Studies on human sexual development. III. Fetal pituitary and serum, and amniotic fluid concentrations of LH, CG, and FSH. J Clin Endocrinol Metab 1976;42:9. Shimoya K, Zhang Q, Tenma K, et al. Fractalkine (FRK) levels in amniotic fluid and its production during pregnancy. Mol Hum Reprod 2003;9:97. Orczyk-Pawilowicz M, Florianski J, Zalewski J, et al. Relative amounts of sialic acid and fucose of amniotic fluid glycoconjugates in relation to pregnancy age. Glycoconj J 2005;22:433. Kvittingen EA, Halvorsen S, Jellum E. Deficient fumarylacetoacetate fumarylhydrolase activity in lymphocytes and fibroblasts from patients with hereditary tyrosinemia. Pediatr Res 1983;17:541. Macek M, Anneren G, Gustavson KH, et al. Gammaglutamyl transferase activity in the amniotic fluid of fetuses with chromosomal aberrations and inborn errors of metabolism. Clin Genet 1987;32:403. Chen AB, Mosesson MW, Solish GI. Identification of the cold-insoluble globulin of plasma in amniotic fluid. Am J Obstet Gynecol 1976;125:958. Newman R, Tutera G. The glucose–insulin ratio in amniotic fluid. Obstet Gynecol 1976;47:599. Papp Z, Ember I, Juhasz E, et al. Acid-soluble glycoproteins in amniotic fluid and cystic fibrosis of the foetus. Clin Genet 1977;11:431. Hakala-Ala-Pietila TH, Koistinen RA, Salonen RK, et al. Elevated second-trimester amniotic fluid concentration of insulin-like growth factor binding protein-1 in fetal growth retardation. Am J Obstet Gynecol 1993;169:35. Appelbaum PC, Shulman G, Chambers NL, et al. Studies on the growth-inhibiting property of amniotic fluids from two United States population groups. Am J Obstet Gynecol 1980;137:579. Jean-Pierre C, Perni SC, Bongiovanni AM, et al. Extracellular 70-kd heat shock protein in mid-trimester amniotic fluid and its effect on cytokine production

CHAPTER 3

265.

266.

267.

268.

269.

270.

271.

272. 273.

274.

275.

276.

277.

Amniotic Fluid Constituents, Cell Culture, and Neural Tube Defects 181

by ex vivo-cultured amniotic fluid cells. Am J Obstet Gynecol 2006;194:694. Gelber SE, Bongiovanni AM, Jean-Pierre C, et al. Witkin SS. Antibodies to the 70 kDa heat shock protein in midtrimester amniotic fluid and intraamniotic immunity. Am J Obstet Gynecol 2007;197:278 e1. Gitlin D, Biasucci A. Development of gamma G, gamma A, gamma M, beta IC-beta IA, C 1 esterase inhibitor, ceruloplasmin, transferrin, hemopexin, haptoglobin, fibrinogen, plasminogen, alpha 1-antitrypsin, orosomucoid, beta-lipoprotein, alpha 2-macroglobulin, and prealbumin in the human conceptus. J Clin Invest 1969;48:1433. Rebmann V, LeMaoult J, Rouas-Freiss N, et al. Quantification and identification of soluble HLA-G isoforms. Tissue Antigens 2007;69 Suppl 1:143. Emmer PM, Steegers EA, van Lierop MJ, et al. Amniotic fluid soluble human leukocyte antigen G is markedly decreased in offspring with neural tube defects. Early Hum Dev 2002;66:101. Deh ME, Klouda PT, Levine M, et al. Detection, isolation and characterization of cell free HLA A and B antigens from human amniotic fluid. Tissue Antigens 1982;20:260. Nicholls T, Hahnel R, Wilkinson S, et al. Identification of 2-hydroxybutyric acid in human amniotic fluid. Clin Chim Acta 1976;69:127. Emery AE, Brock DJ, Burt D, et al. Amniotic fluid composition in malformations of the fetal central nervous system. J Obstet Gynaecol Br Commonw 1974;81:512. GadowEC, Floriani FA, Florin A. IgG levels in amniotic fluid. Am J Obstet Gynecol 1974;119:849. Wilfert CM, Gradoville ML. In vitro viral studies of cells grown from human amniotic fluid samples. Am J Obstet Gynecol 1974;118:1073. Thirunavukarasu PP, Lambert-Messerlian G, Robertson DM, et al. Molecular weight forms of inhibin A, inhibin B and pro-alpha C in maternal serum, amniotic fluid and placental extracts of normal and Down syndrome pregnancies. Prenat Diagn 2002;22:1086. Verhaeghe J, Coopmans W, van Herck E, et al. IGF-I, IGF-II, IGF binding protein 1, and C-peptide in second trimester amniotic fluid are dependent on gestational age but do not predict weight at birth. Pediatr Res 1999;46:101. Weiss PA, Purstner P, Winter R, et al. Insulin levels in amniotic fluid of normal and abnormal pregnancies. Obstet Gynecol 1984;63:371. Baviera G, D’Anna R, Corrado F, et al. ICAM-1 in maternal serum and amniotic fluid as an early marker of preeclampsia and IUGR. J Reprod Med 2002;47:191.

278. Malamitsi-Puchner A, Vrachnis N, Samoli E, et al. Elevated second trimester amniotic fluid interferon gamma-inducible T-cell alpha chemoattractant concentrations as a possible predictor of preterm birth. J Soc Gynecol Investig 2006;13:25. 279. Menon R, Camargo MC, Thorsen P, et al. Amniotic fluid interleukin-6 increase is an indicator of spontaneous preterm birth in white but not black Americans. Am J Obstet Gynecol 2008;198:77 e1. 280. Rayburn WF, Robinson A, Braverman LE, et al. Iodide concentrations in matched maternal serum, cord serum, and amniotic fluid from preterm and term human pregnancies. Reprod Toxicol 2008;25:129. 281. Garcia-Fuentes E, Gallo M, Garcia L, et al. Amniotic fluid iodine concentrations do not vary in pregnant women with varying iodine intake. Br J Nutr 2008;99:1178. 282. Maymon R, Jauniaux E, Rodeck C, et al. Comparison of placental isoferritin levels in maternal serum and coelomic and amniotic fluids during first trimester human gestation. Hum Reprod 1998;13:1044. 283. Wolf RO, Taussig LM. Human amniotic fluid isoamylases. Functional development of fetal pancreas and salivary glands. Obstet Gynecol 1973;41:337. 284. Perrone S, Longini M, Bellieni CV, et al. Early oxidative stress in amniotic fluid of pregnancies with Down syndrome. Clin Biochem 2007;40:177. 285. Shaw JL, Diamandis EP. Distribution of 15 human kallikreins in tissues and biological fluids. Clin Chem 2007;53:1423. 286. Kidokoro K, Furuhashi M, Kuno N, et al. Amniotic fluid neutrophil elastase and lactate dehydrogenase: association with histologic chorioamnionitis. Acta Obstet Gynecol Scand 2006;85:669. 287. Otsuki K, Yoda A, Saito H, et al. Amniotic fluid lactoferrin in intrauterine infection. Placenta 1999;20:175. 288. Stoll C, Dott B, Maier EA, et al. Oligo-éléments du liquide amniotique des foetus normaux, hypotrophes et trisomiques 21 [Oligo-elements of the amniotic fluid from normal, hypotrophic and trisomy 21 fetuses]. Rev Fr Gynecol Obstet 1990;85:45. 289. Larsen B, Galask RP, Snyder IS. Muramidase and peroxidase activity of human amniotic fluid. Obstet Gynecol 1974;44:219. 290. Hayashi M, Zhu K, Sagesaka T, et al. Elevation of amniotic fluid macrophage colony-stimulating factor in normotensive pregnancies that delivered small-for-gestational-age infants. Am J Reprod Immunol 2007;57:488. 291. Fujikura T, Klionsky B. The significance of meconium staining. Am J Obstet Gynecol 1975;121:45.

182 Genetic Disorders and the Fetus

292. Kim KW, Romero R, Park HS, et al. A rapid matrix metalloproteinase-8 bedside test for the detection of intraamniotic inflammation in women with preterm premature rupture of membranes. Am J Obstet Gynecol 2007;197:292 e1. 293. Fasching G, Haeusler M, Mayr J, et al. Can levels of interleukins and matrix metalloproteinases in the amniotic fluid predict postnatal bowel function in fetuses with gastroschisis? J Pediatr Surg 2005;40: 1887. 294. Nishijima K, Shukunami K, Tsukahara H, et al. Micelles of pulmonary surfactant in human amniotic fluid at term. Pediatr Res 2006;60:196. 295. Cassell GH, Davis RO, Waites KB, et al. Isolation of Mycoplasma hominis and Ureaplasma urealyticum from amniotic fluid at 16–20 weeks of gestation: potential effect on outcome of pregnancy. Sex Transm Dis 1983;10:294. 296. Kastner B, Schenk H, Weise W, et al. Der Neuraminsaure/Protein-Quotient im Fruchtwasser: Indikator fur die fetale Reife [The neuraminic acid/protein quotient in amniotic fluid: indicator for fetal maturity]. Z Med Lab Diagn 1990;31:77. 297. Marx CE, Vance BJ, Jarskog LF, et al. Nerve growth factor, brain-derived neurotrophic factor, and neurotrophin-3 levels in human amniotic fluid. Am J Obstet Gynecol 1999;181:1225. 298. Elimian A, Figueroa R, Patel K, et al. Reference values of amniotic fluid neuron-specific enolase. J Matern Fetal Med 2001;10:155. 299. Daoud GA, Suzuki Y,Yamamoto T, et al. Reduced nitric oxide in amniotic fluid of patients with chorioamnionitis. Fetal Diagn Ther 2006;21:77. 300. Tranquilli AL, Bezzeccheri V, Scagnoli C, et al. Amniotic levels of vascular endothelial growth factor and nitric oxide at the second trimester in Down’s syndrome. J Matern Fetal Neonatal Med 2003;13:28. 301. Dawson EB, Harris WA, Evans DR, et al. Amniotic fluid amino and nucleic acid in normal and neural tube defect pregnancies. A comparison. J Reprod Med 1999;44:28. 302. Lu LC, Hsu CD. Elevated amniotic fluid nucleosome levels in women with intra-amniotic infection. Obstet Gynecol 1999;94:7. 303. Ruhle W, Graf von Ballestrem CL, Pult HM, et al. Korrelation des Cotininspiegels in Fruchtwasser, Nabelarterienblut und mutterlichem Blut [Correlation of cotinine level in amniotic fluid, umbilical artery blood and maternal blood]. Geburtshilfe Frauenheilkd 1995;55:156. 304. Ramsay SL, Maire I, Bindloss C, et al. Determination of oligosaccharides and glycolipids in amniotic fluid by electrospray ionisation tandem mass spectrometry:

305. 306.

307. 308.

309.

310.

311.

312.

313.

314.

315.

316.

317.

318.

in utero indicators of lysosomal storage diseases. Mol Genet Metab 2004;83:231. Nicholls TM, Hahnel R, Wilkinson SP. Organic acids in amniotic fluid. Clin Chim Acta 1978;84:11. Baggot PJ, Eliseo AJ, DeNicola NG, et al. Organic acid concentrations in amniotic fluid found in normal and Down syndrome pregnancies. Fetal Diagn Ther 2008;23:245. Mattison DR. Amniotic fluid osmolality. Obstet Gynecol 1970;36:420. Wandzilak TR, Hanson FW, Williams HE. The quantitation of oxalate in amniotic fluid by ion chromatography. Clin Chim Acta 1989;185:131. Johnell HE, Nilsson BA. Oxygen tension, acid-base status and electrolytes in human amniotic fluid. Acta Obstet Gynecol Scand 1971;50:183. Seppala M, Aho I, Tissari A, et al. Radioimmunoassay of oxytocin in amniotic fluid, fetal urine, and meconium during late pregnancy and delivery. Am J Obstet Gynecol 1972;114:788. Westergaard JG, Chemnitz J, Teisner B, et al. Pregnancy-associated plasma protein A: a possible marker in the classification and prenatal diagnosis of Cornelia de Lange syndrome. Prenat Diagn 1983;3:225. Schlievert P, Johnson W, Galask RP. Bacterial growth inhibition by amniotic fluid. V. Phosphate-to-zinc ratio as a predictor of bacterial growth-inhibitory activity. Am J Obstet Gynecol 1976;125:899. Mihailovic M, Cvetkovic M, Ljubic A, et al. Selenium and malondialdehyde content and glutathione peroxidase activity in maternal and umbilical cord blood and amniotic fluid. Biol Trace Elem Res 2000;73:47. Condroelli S, Cosmi EV, Scarpelli EM. Extrapulmonary source of amniotic fluid phospholipids. Am J Obstet Gynecol 1974;118:842. Moon TC, Lee JH, Lee SH, et al. Detection and characterization of a type IIA secretory phospholipase A2 inhibitory protein in human amniotic fluid. Biol Pharm Bull 2000;23:1163. Foster WG, Chan S, Platt L, Hughes CL, Jr. Detection of phytoestrogens in samples of second trimester human amniotic fluid. Toxicol Lett 2002;129:199. Makrydimas G, Sotiriadis A, Savvidou MD, et al. Physiological distribution of placental growth factor and soluble Flt-1 in early pregnancy. Prenat Diagn 2008;28:175. Vesce F, Scapoli C, Giovannini G, et al. Plasminogen activator system in serum and amniotic fluid of euploid and aneuploid pregnancies. Obstet Gynecol 2001;97:404.

CHAPTER 3

Amniotic Fluid Constituents, Cell Culture, and Neural Tube Defects 183

319. Verkleij-Hagoort AC, Ursem NT, Hop WC, et al. Complex congenital malformations and the impact of the plasminogen activator system and beta-hCG in amniotic fluid. Eur J Obstet Gynecol Reprod Biol 2007;135:47. 320. Prado VF, Reis DD, Pena SD. Biochemical and immunochemical identification of the fetal polypeptides of human amniotic fluid during the second trimester of pregnancy. Braz J Med Biol Res 1990;23:121. 321. Phillips LL, Davidson EC, Jr. Procoagulant properties of amniotic fluid. Am J Obstet Gynecol 1972;113: 911. 322. Kauppila S, Tekay A, Risteli L, et al. Type I and type III procollagen propeptides in amniotic fluid of normal pregnancies and in a case of mild osteogenesis imperfecta. Eur J Clin Invest 1998;28:831. 323. Ben-Jonathan N, Munsick RA. Dopamine and prolactin in human pregnancy. J Clin Endocrinol Metab 1980;51:1019. 324. Singh EJ, Zuspan FP. Content of amniotic fluid prostaglandins in normal, diabetic, and drug-abuse human pregnancy. Am J Obstet Gynecol 1974;118: 358. 325. Graca G, Duarte IF, Goodfellow BJ, et al. Potential of NMR spectroscopy for the study of human amniotic fluid. Anal Chem 2007;79:8367. 326. Kaufman S. Protein-bound iodine (pbi) in human amniotic fluid. J Pediatr 1966;68:990. 327. Uszynski M, Zekanowska E, Kotzbach M, et al. Protein C, protein S, and thrombomodulin in amniotic fluid. A preliminary study. J Perinat Med 2006;34:289. 328. Chi L, Wolff JJ, Laremore TN, et al. Structural analysis of bikunin glycosaminoglycan. J Am Chem Soc 2008;130:2617. 329. Porre S, Heinonen S, Mantyjarvi R, et al. Psoriasin, a calcium-binding protein with chemotactic properties is present in the third trimester amniotic fluid. Mol Hum Reprod 2005;11:87. 330. Franks RC, Hayashi RH. Maternal and fetal renin activity and renin and big renin concentrations in second-trimester pregnancy. Am J Obstet Gynecol 1979;134:20. 331. Cederqvist LL, Zervoudakis IA, Ewool LC, et al. Prenatal diagnosis of congenital rubella. Br Med J 1977;1:615. 332. Teichler-Zallen D, Doherty RA. Amniotic fluid secretor typing: validation for use in prenatal prediction of myotonic dystrophy. Clin Genet 1980;18:257. 333. Aula N, Aula P. Prenatal diagnosis of free sialic acid storage disorders (SASD). Prenat Diagn 2006;26:655.

334. Cox LW, Chalmers TA. The transfer of sodium to the amniotic fluid in normal and abnormal cases, determined by Na24 tracer methods. J Obstet Gynaecol Br Emp 1953;60:222. 335. Tham A, Wetterberg L, Sara VR. Immunoreactive somatomedin B in the human foetus and in women during pregnancy. Acta Endocrinol (Copenh) 1987;115:218. 336. Niswander KR. Spectrophotometric analysis of amniotic fluid in early gestation. Am J Obstet Gynecol 1970;108:1296. 337. Fortunato SJ, Menon R, Lombardi SJ. Stromelysins in placental membranes and amniotic fluid with premature rupture of membranes. Obstet Gynecol 1999;94:435. 338. Frosolono MF, Roux JF. Surface-active material in human amniotic fluid. Am J Obstet Gynecol 1978;130: 562. 339. Muller-Tyl E, Lempert J, Steinbereithner K, et al. Surface properties of the amniotic fluid in normal pregnancy. Am J Obstet Gynecol 1975;122:295. 340. Belisle S, Fencl MM, Tulchinsky D. Amniotic fluid testosterone and follicle-stimulating hormone in the determination of fetal sex. Am J Obstet Gynecol 1977;128:514. 341. Tersenov OA, Mikhaleva IV, Usol’tseva VA, et al. O svertyvaiushchei aktivnosti okoloplodnykh vod [Coagulative activity of the amniotic fluid]. Akush Ginekol (Mosk) 1989:43. 342. Sainio S, Javela K, Kekomaki R, et al. Thrombopoietin levels in cord blood plasma and amniotic fluid in fetuses with alloimmune thrombocytopenia and healthy controls. Br J Haematol 2000;109:330. 343. Hall GS, Carr MJ, Cummings E, et al. Aluminum, barium, silicon, and strontium in amniotic fluid by emission spectrometry. Clin Chem 1983;29:1318. 344. Hufner M, Grussendorf M, Lorenz U, et al. 3,3′ ,5′ -Triiodothyronine (reverse T3) in amniotic fluid and cord serum. Eur J Pediatr 1977;125:213. 345. Stefanovic V, Loukovaara M. Amniotic fluid cardiac troponin T in pathological pregnancies with evidence of chronic fetal hypoxia. Croat Med J 2005;46: 801. 346. Menon R, Thorsen P, Vogel I, et al. Racial disparity in amniotic fluid concentrations of tumor necrosis factor (TNF)-alpha and soluble TNF receptors in spontaneous preterm birth. Am J Obstet Gynecol 2008;198: 533 e1. 347. Park CW, Park JS, Shim SS, et al. An elevated maternal plasma, but not amniotic fluid, soluble fms-like tyrosine kinase-1 (sFlt-1) at the time of mid-trimester genetic amniocentesis is a risk factor for preeclampsia. Am J Obstet Gynecol 2005;193:984.

184 Genetic Disorders and the Fetus

348. Enlander D. Amniotic fluid indicators of fetal maturity. Obstet Gynecol 1972;40:605. 349. Kobayashi H, Suzuki K, Sugino D, et al. Urinary trypsin inhibitor levels in amniotic fluid of normal human pregnancy: decreased levels observed at parturition. Am J Obstet Gynecol 1999;180:141. 350. Wallingford JC, Milunsky A, Underwood BA. Vitamin A and retinol-binding protein in amniotic fluid. Am J Clin Nutr 1983;38:377. 351. Gardiki-Kouidou P, Seller MJ. Amniotic fluid folate, vitamin B12 and transcobalamins in neural tube defects. Clin Genet 1988;33:441. 352. Weisman Y, Jaccard N, Legum C, et al. Prenatal diagnosis of vitamin D-dependent rickets, type II: response to 1,25-dihydroxyvitamin D in amniotic fluid cells and fetal tissues. J Clin Endocrinol Metab 1990;71:937. 353. Lacourt GC, Arendt J, Cox J, et al. Microcephalic dwarfism with associated low amniotic fluid 5-hydroxyindole-3-acetic acid (5HIAA). Report of a case of Cornelia de Lange syndrome. Helv Paediatr Acta 1977;32:149. 354. Weiss PA, Kainer F, Haeusler M, et al. Amniotic fluid insulin levels in nondiabetic pregnant women: an update. Arch Gynecol Obstet 1998;262:81. 355. Johnson MR, Abbas A, Nicolaides KH, et al. Distribution of relaxin between human maternal and fetal circulations and amniotic fluid. J Endocrinol 1992;134:313. 356. Caserta D, Mantovani A, Ciardo F, et al. Heavy metals in human amniotic fluid: a pilot study. Prenat Diagn 2011;31:792. 357. Caserta D, Graziano A, Lo Monte G, et al. Heavy metals and placental fetal-maternal barrier: a mini-review on the major concerns. Eur Rev Med Pharmacol Sci 2013;17:2198. 358. Chez RA, Henkin RI, Fox R. Amniotic fluid copper and zinc concentrations in human pregnancy. Obstet Gynecol 1978;52:125. 359. Nusbaum MJ, Zettner A. The content of calcium, magnesium, copper, iron, sodium, and potassium in amniotic fluid from eleven to nineteen weeks’ gestation. Am J Obstet Gynecol 1973;115:219. 360. Keppen LD, Pysher T, Rennert OM. Zinc deficiency acts as a co-teratogen with alcohol in fetal alcohol syndrome. Pediatr Res 1985;19:944. 361. Ward N, Bryce-Smith DM, Minski M, et al. Multi element neutron activation analysis of amniotic fluid in relation to varying gestational membrane ruptures. In: Bratter P, Schramel P, eds. Analytical chemistry in medicine and biology. Proceedings of the 2nd International Workshops. Berlin: de Gruyter, 1983:483.

362. Parkinson CE, Tan JC, Gal I. Vitamin A concentration in amniotic fluid and maternal serum related to neural tube defects. Br J Obstet Gynaecol 1982;89:935. 363. Parkinson CE, Tan JC, Lewis PJ, et al. Amniotic fluid zinc and copper and neural tube defects. J Obstet Gynaecol 1981;1:207. 364. Tamura T, Weekes EW, Birch R, et al. Relationship between amniotic fluid and maternal blood nutrient levels. J Perinat Med 1994;22:227. 365. Dawson EB, Evans DR, Van Hook JW. Amniotic fluid B12 and folate levels associated with neural tube defects. Am J Perinatol 1998;15:511. 366. Dawson EB, Evans DR, Harris WA, et al. Amniotic fluid B12, calcium, and lead levels associated with neural tube defects. Am J Perinatol 1999;16: 373. 367. Luglie PF, Frulio A, Campus G, et al. Dosaggio del mercurio nel liquido amniotico umano [Mercury determination in human amniotic fluid]. Minerva Stomatol 2000;49:155. 368. Kozikowska I, Binkowski LJ, Szczepanska K, et al. Mercury concentrations in human placenta, umbilical cord, cord blood and amniotic fluid and their relations with body parameters of newborns. Environ Pollut 2013;182:256. 369. Milnerowicz H, Zalewski J, Geneja R, et al. Poziom metali Cd i Pb we krwi oraz Zn, Cu, Cd i Pb w wodach płodowych kobiet palacych papierosy podczas ciazy powikłanej małowodziem lub przedwczesnym odpływaniem w’od płodowych [Levels of Cd, Pb in blood and Zn, Cu, Cd, Pb in amniotic fluid of tobacco smoking women during pregnancy complicated oligohydramnios or premature rupture of membranes]. Ginekol Pol 2000;71:311. 370. Hodari AA, Mariona FG, Houlihan RT, et al. Creatinine transport in the maternal–fetal complex. Obstet Gynecol 1973;41:47. 371. Droegemueller W, Jackson C, Makowski EL, et al. Amniotic fluid examination as an aid in the assessment of gestational age. Am J Obstet Gynecol 1969;104: 424. 372. Sharma SD, Trussell RR. The value of amniotic fluid examination in the assessment of fetal maturity. J Obstet Gynaecol Br Commonw 1970;77:215. 373. Muller F, Bernard MA, Benkirane A, et al. Fetal urine cystatin C as a predictor of postnatal renal function in bilateral uropathies. Clin Chem 1999;45:2292. 374. Renwick JH, Lawler SD. Genetical linkage between the ABO and nail-patella loci. Ann Hum Genet 1955;19:312. 375. Bongers EM, de Wijs IJ, Marcelis C, et al. Identification of entire LMX1B gene deletions in nail patella syndrome: evidence for haploinsufficiency as the main

CHAPTER 3

376.

377.

378.

379.

380.

381.

382.

383.

384.

385. 386.

387.

388.

389. 390. 391.

Amniotic Fluid Constituents, Cell Culture, and Neural Tube Defects 185

pathogenic mechanism underlying dominant inheritance in man. Eur J Hum Genet 2008;16:1240. Renwick JH, Bundey SE, Ferguson-Smith MA, Izatt MM. Confirmation of linkage of the loci for myotonic dystrophy and ABH secretion. J Med Genet 1971;8:407. Milunsky A, Skare JC, Milunsky JM, et al. Prenatal diagnosis of myotonic muscular dystrophy with linked deoxyribonucleic acid probes. Am J Obstet Gynecol 1991;164:751. Kamsteeg EJ, Kress W, Catalli C, et al. Best practice guidelines and recommendations on the molecular diagnosis of myotonic dystrophy types 1 and 2. Eur J Hum Genet 2012;20:1203. Durandy A, Cerf-Bensussan N, Dumez Y, et al. Prenatal diagnosis of severe combined immunodeficiency with defective synthesis of HLA molecules. Prenat Diagn 1987;7:27. Dundar O, Yoruk P, Tutuncu L, et al. Second trimester amniotic fluid annexin A5 levels and subsequent development of intrauterine growth restriction. Prenat Diagn 2008;28:887. Davis LE, McLaren LC, Stewart JA, et al. Immunological and microbiological studies of midtrimester amniotic fluid. Gynecol Obstet Invest 1983;16:261. Boulley AM, Vial M, Bessis R, et al. Diagnostic antenatal de deux cas de maladie des inclusions cytomegaliques [Antenatal diagnosis of 2 cases of cytomegalic inclusion disease]. Arch Fr Pediatr 1984;41:123. Auger P, Marquis G, Dallaire L, et al. Natural occurrence of a humoral response to Candida in human amniotic fluid. Am J Obstet Gynecol 1980;136:1075. Emelyanova AI, Mikheeva GA, Ermakova GG. Amniotic fluid immunoglobulins at different dates of pregnancy [in Russian]. Akush Ginekol (Mosk) 1982;7:19. Harwick HJ, Iuppa JB, Fekety FR, Jr. Microorganisms and amniotic fluid. Obstet Gynecol 1969;33:256. Tafari N, Ross SM, Naeye RL, et al. Failure of bacterial growth inhibition by amniotic fluid. Am J Obstet Gynecol 1977;128:187. Thadepalli H, Appleman MD, Maidman JE, et al. Antimicrobial effect of amniotic fluid against anaerobic bacteria. Am J Obstet Gynecol 1977;127:250. Walsh H, Hildebrandt RJ, Prystowsky H. Growth inhibition factors in amniotic fluid. Am J Obstet Gynecol 1965;93:590. Sarkany I, Gaylarde CC. The effect of amniotic fluid on bacterial growth. Br J Dermatol 1968;80:241. Gusdon JP. A bactericidin for Bacillus subtilis in pregnancy. J Immunol 1962;88:494. Galask RP, Snyder IS. Bacterial inhibition by amniotic fluid. Am J Obstet Gynecol 1968;102:949.

392. Altieri C, Maruotti G, Natale C, et al. In vitro survival of Listeria monocytogenes in human amniotic fluid. Zentralbl Hyg Umweltmed 1999;202:377. 393. Bratlid D, Lindback T. Bacteriolytic activity of amniotic fluid. Obstet Gynecol 1978;51:63. 394. Schlievert PS, Johnson W, Galask RP. Bacterial growth inhibition by amniotic fluid. VI. Evidence for a zinc peptide antibacterial system. Am J Obstet Gynecol 1976;125:906. 395. Soto E, Espinoza J, Nien JK, et al. Human beta-defensin-2: a natural antimicrobial peptide present in amniotic fluid participates in the host response to microbial invasion of the amniotic cavity. J Matern Fetal Neonatal Med 2007;20:15. 396. Thadepalli H, Bach VT, Davidson EC, Jr. Antimicrobial effect of amniotic fluid. Obstet Gynecol 1978;52:198. 397. Kim JY, Park SC, Lee JK, et al. Novel antibacterial activity of beta(2)-microglobulin in human amniotic fluid. PLoS One 2012;7:e47642. 398. Evaldson G, Nord CE. Amniotic fluid activity against Bacteroides fragilis and group B streptococci. Med Microbiol Immunol 1981;170:11. 399. Ismail MA, Salti GI, Moawad AH. Effect of amniotic fluid on bacterial recovery and growth: clinical implications. Obstet Gynecol Surv 1989;44:571. 400. Martius J, Eschenbach DA. The role of bacterial vaginosis as a cause of amniotic fluid infection, chorioamnionitis and prematurity – a review. Arch Gynecol Obstet 1990;247:1. 401. Farzin A, Boyer P, Ank B, et al. Amniotic fluid exhibits an innate inhibitory activity against HIV type 1 replication in vitro. AIDS Res Hum Retroviruses 2013;29:77. 402. Charles D, Edwards WR. Infectious complications of cervical cerclage. Am J Obstet Gynecol 1981;141:1065. 403. Lebon P, Girard S, Thepot F, et al. Embryologie générale: présence constante d’interféron de type a dans les liquides amniotiques humains. C R Hebd Seances Acad Sci Paris 1981;293:69. 404. Morgan-Capner P, Rodeck CH, Nicolaides KH, et al. Prenatal detection of rubella-specific IgM in fetal sera. Prenat Diagn 1985;5:21. 405. Ho-Terry L, Terry GM, Londesborough P. Diagnosis of foetal rubella virus infection by polymerase chain reaction. J Gen Virol 1990;71 (Pt 7):1607. 406. Bosma TJ, Corbett KM, Eckstein MB, et al. Use of PCR for prenatal and postnatal diagnosis of congenital rubella. J Clin Microbiol 1995;33:2881. 407. Combs CA, Gravett M, Garite TJ, et al. Amniotic fluid infection, inflammation, and colonization in preterm labor with intact membranes. Am J Obstet Gynecol 2014;210:125 e1.

186 Genetic Disorders and the Fetus

408. Ardissone AN, de la Cruz DM, Davis-Richardson AG, et al. Meconium microbiome analysis identifies bacteria correlated with premature birth. PLoS One 2014;9:e90784. 409. Pons JC, Vial P, Rozenberg F, et al. Diagnostic prenatal de la foetopathie varicelleuse au deuxi‘eme trimester de la grossesse. [Prenatal diagnosis of fetal varicella in the second trimester of pregnancy]. J Gynecol Obstet Biol Reprod (Paris) 1995;24:829. 410. Dufour P, de Bievre P, Vinatier D, et al. Varicella and pregnancy. Eur J Obstet Gynecol Reprod Biol 1996;66:119. 411. Shukla SK, Meier PR, Mitchell PD, et al. Leptotrichia amnionii sp. nov., a novel bacterium isolated from the amniotic fluid of a woman after intrauterine fetal demise. J Clin Microbiol 2002;40:3346. 412. Blanco JD, Gibbs RS, Krebs LF, et al. The association between the absence of amniotic fluid bacterial inhibitory activity and intra-amniotic infection. Am J Obstet Gynecol 1982;143:749. 413. Grose C, Weiner CP. Prenatal diagnosis of congenital cytomegalovirus infection: two decades later. Am J Obstet Gynecol 1990;163:447. 414. Derouin F, Thulliez P, Candolfi E, et al. Early prenatal diagnosis of congenital toxoplasmosis using amniotic fluid samples and tissue culture. Eur J Clin Microbiol Infect Dis 1988;7:423. 415. Leiberman JR, Hagay ZJ, Dagan R. Intraamniotic Haemophilus influenzae infection. Arch Gynecol Obstet 1989;244:183. 416. Zauli DA, de Menezes CL, de Oliveira CL. Development and padronization of three multiplex PCRs for the diagnosis of Chlamydia trachomatis, Toxoplasma gondii, herpes simplex viruses 1 and 2, and cytomegalovirus. Mol Biotechnol 2013;54:1004. 417. Curtin WM, Menegus MA, Patru MM, et al. Midtrimester fetal herpes simplex-2 diagnosis by serology, culture and quantitative polymerase chain reaction. Fetal Diagn Ther 2013;33:133. 418. Czader M, Post K, Cheng L. Detection of cytomegalovirus infection by quantitative polymerase chain reaction. Methods Mol Biol 2013;999:257. 419. Bourgeois-Nicolaos N, Cordier AG, Guillet-Caruba C, et al. Evaluation of the Cepheid Xpert GBS assay for rapid detection of group B streptococci in amniotic fluids from pregnant women with premature rupture of membranes. J Clin Microbiol 2013;51:1305. 420. Bernard B, Barton L, Abate M, et al. Maternal–fetal transfer of cefazolin in the first twenty weeks of pregnancy. J Infect Dis 1977;136:377.

421. Giamarellou H, Gazis J, Petrikkos G, et al. A study of cefoxitin, moxalactam, and ceftazidime kinetics in pregnancy. Am J Obstet Gynecol 1983;147:914. 422. Allegaert K, van Mieghem T, Verbesselt R, et al. Cefazolin pharmacokinetics in maternal plasma and amniotic fluid during pregnancy. Am J Obstet Gynecol 2009;200:170 e1. 423. Atkinson G, Campbell DJ, Cawood ML, et al. Steroids in human intrauterine fluids of early pregnancy. Clin Endocrinol (Oxf) 1996;44:435. 424. Nakanishi T, Suzumori N, Mizuno H, et al. Elevated aldosterone in amniotic fluid and maternal blood has diagnostic potential in pregnancies complicated with a fetus of Bartter syndrome. Fetal Diagn Ther 2005;20:481. 425. Mennuti MT, Wu CH, Mellman WJ, et al. Amniotic fluid testosterone and follicle stimulating hormone levels as indicators of fetal sex during mid-pregnancy. Am J Med Genet 1977;1:211. 426. Wisniewski L, Jezuita J, Bogoniowska Z, et al. Endokrynologia prenatalna. I. Kortyzol w plynie owodniowym i w surowicy krwi kobiet w 17 tygodniu ciazy [Prenatal endocrinology. II. Growth hormone in the amniotic fluid and blood serum of women in the 17th week of pregnancy]. Ginekol Pol 1983;54:693. 427. Goebelsmann U, Abboud TK, Hoffman DI, et al. Beta endorphin in pregnancy. Eur J Obstet Gynecol Reprod Biol 1984;17:77. 428. Teramo K, Kari MA, Eronen M, et al. High amniotic fluid erythropoietin levels are associated with an increased frequency of fetal and neonatal morbidity in type 1 diabetic pregnancies. Diabetologia 2004;47:1695. 429. Freeman R, Lev-Gur M, Koslowe R, et al. Maternal plasma and amniotic fluid levels of estradiol, estrone, progesterone, and prolactin in early pregnancy. Obstet Gynecol 1984;63:507. 430. Sugar J, Dessy C, Alexander S, et al. Estriol-3-glucuronide and estriol-16-glucuronide in amniotic fluid during normal pregnancy. J Clin Endocrinol Metab 1980;50:137. 431. Zamlynski J, Chudek J, Olejek A, et al. Galanin concentrations in maternal circulation, amniotic fluid and umbilical cord blood during term labor: relationship with maternal body mass and neonatal birth weight. Gynecol Endocrinol 2007;23:295. 432. Heikinheimo M, Seppala M, Brock DJ. Pregnancy specific beta 1-glycoprotein and human chorionic gonadotrophin levels in amniotic fluid and maternal serum in the first half of pregnancy. Oncodev Biol Med 1980;1:71.

CHAPTER 3

Amniotic Fluid Constituents, Cell Culture, and Neural Tube Defects 187

433. Dattatreyamurty B, Sheth AR, Purandare TV, et al. Gonadotrophins during second trimester of pregnancy: I. LH and hCG levels in maternal serum and amniotic fluid and their relationship to the sex of the foetus. Acta Endocrinol (Copenh) 1979;91:692. 434. Buhling KJ, Monnich J, Henrich W, et al. Zusammenhang zwischen Fruchtwasserinsulin im zweiten Trimenon, mutterlicher Glukosetoleranz und fetalen Fehlbildungen [Association between amniotic fluid insulin in the second trimester, maternal glucose tolerance and fetalmalformations]. Z Geburtshilfe Neonatol 2004;208:226. 435. Tisi DK, Liu XJ, Wykes LJ, et al. Insulin-like growth factor II and binding proteins 1 and 3 from second trimester human amniotic fluid are associated with infant birth weight. J Nutr 2005;135:1667. 436. Bay K, Cohen AS, Jorgensen FS, et al. Insulin-like factor 3 levels in second-trimester amniotic fluid. J Clin Endocrinol Metab 2008;93:4048. 437. D’Anna R, Baviera G, Cannata ML, et al. Midtrimester amniotic fluid leptin and insulin levels and subsequent gestational diabetes. Gynecol Obstet Invest 2007;64:65. 438. Marek S, Bock K, Hellmeyer L, et al. Herkunft und Bedeutung von Leptin und Neuropeptid Y (NPY) im Fruchtwasser zwischen der 14.- 18. Schwangerschaftswoche [Origin and significance of leptin and neuropeptide Y (NPY) in amniotic fluid between the 14th and 18th weeks of gestation]. Z Geburtshilfe Neonatol 2006;210:208. 439. Petrucha RA, Goebelsmann U, Hung TT, et al. Amniotic fluid beta-endorphin and beta-lipotropin concentrations during the second and third trimesters. Am J Obstet Gynecol 1983;146:644. 440. Shi-hao Y. Prolaktin u amnijskoj tekucini i serumu u ranoj trudnoci. [Prolactin in amnionic fluid and serum of women in early pregnancy (author’s transl.)]. Jugosl Ginekol Opstet 1981;21:116. 441. Lee SE, Romero R, Park IS, et al. Amniotic fluid prostaglandin concentrations increase before the onset of spontaneous labor at term. J Matern Fetal Neonatal Med 2008;21:89. 442. van de Beek C, Thijssen JH, Cohen-Kettenis PT, et al. Relationships between sex hormones assessed in amniotic fluid, and maternal and umbilical cord serum: what is the best source of information to investigate the effects of fetal hormonal exposure? Horm Behav 2004;46:663. 443. Cooper E, Anderson A, Bennett MJ, et al. Radioimmunoassay of thyroxine and 3,3′ ,5′ -triiodothyronine (reverse T3) in human amniotic fluid. Clin Chim Acta 1982;118:57.

444. Vascotto C, Salzano AM, D’Ambrosio C, et al. Oxidized transthyretin in amniotic fluid as an early marker of preeclampsia. J Proteome Res 2007;6:160. 445. Wu CH, Mennuti MT, Mikhail G. Free and protein-bound steroids in amniotic fluid of midpregnancy. Am J Obstet Gynecol 1979;133:666. 446. Merger C, Blanc B, Ruf H, et al. Dosage de la testosterone, delta 4-androstenedione et S-DHA dans le liquide amniotique et sexe foetal [The level of testosterone, delta 4-androstenedione, and S-DHA in liquor and fetal sex (author’s transl.)]. J Gynecol Obstet Biol Reprod (Paris) 1981;10:567. 447. Carson DJ, Okuno A, Lee PA, et al. Amniotic fluid steroid levels. Fetuses with adrenal hyperplasia, 46,XXY fetuses, and normal fetuses. Am J Dis Child 1982;136:218. 448. Mean F, Pescia G, Vajda D, et al. Amniotic fluid testosterone in prenatal sex determination. J Genet Hum 1981;29:441. 449. Robinson JD, Judd HL, Young PE, et al. Amniotic fluid androgens and estrogens in midgestation. J Clin Endocrinol Metab 1977;45:755. 450. Abramovich DR, Herriot R, Stott J. Dihydrotestosterone levels at midpregnancy and term: a comparison with testosterone concentrations. Br J Obstet Gynaecol 1983;90:232. 451. Perera DM, McGarrigle HH, Lawrence DM, et al. Amniotic fluid testosterone and testosterone glucuronide levels in the determination of foetal sex. J Steroid Biochem 1987;26:273. 452. Roig MD, Sabria J, Valls C, et al. The use of biochemical markers in prenatal diagnosis of intrauterine growth retardation: insulin-like growth factor I, Leptin, and alpha-fetoprotein. Eur J Obstet Gynecol Reprod Biol 2005;120:27. 453. Anand-Ivell R, Ivell R, Driscoll D, et al. Insulin-like factor 3 levels in amniotic fluid of human male fetuses. Hum Reprod 2008;23:1180. 454. Blankstein J, Kraiem Z, Mashiah S, et al. Effect of dexamethasone administration at midgestation on cortisol levels in amniotic fluid. Isr J Med Sci 1979;15:945. 455. Friedrich E, Habedank M, Cooreman G, et al. Diagnostik der fotalen Schilddrusenfunktion durch Amniocentese [Diagnostic possibilities of the function of the fetal thyroid gland by amniocentesis (author’s transl.)]. Z Geburtshilfe Perinatol 1981;185:96. 456. Perelman AH, Johnson RL, Clemons RD, et al. Intrauterine diagnosis and treatment of fetal goitrous hypothyroidism. J Clin Endocrinol Metab 1990;71:618. 457. Stoll C, Willard D, Czernichow P, et al. Prenatal diagnosis of primary pituitary dysgenesis. Lancet 1978;i:932.

188 Genetic Disorders and the Fetus

458. Buscher U, Hertwig K, Dudenhausen JW. Nachweis von Erythropoietin im Fruchtwasser [Detection of erythropoietin in amniotic fluid]. Geburtshilfe Frauenheilkd 1996;56:243. 459. Kakuya F, Shirai M, Takase M, et al. Relationship between erythropoietin levels both in cord serum and amniotic fluid at birth and abnormal fetal heart rate records. Pediatr Int 2002;44:414. 460. Teramo KA, Widness JA. Increased fetal plasma and amniotic fluid erythropoietin concentrations: markers of intrauterine hypoxia. Neonatology 2009;95:105. 461. Mikovic Z, Mandic V, Parovic V, et al. Erythropoietin in amniotic fluid as a potential marker in distinction between growth restricted and constitutionally small fetuses. J Matern Fetal Neonatal Med 2014;27:1134. 462. Basbug M, Serin IS, Ozcelik B, et al. Correlation of elevated leptin levels in amniotic fluid and maternal serum in neural tube defects. Obstet Gynecol 2003;101:523. 463. Bala RM, Wright C, Bardai A, et al. Somatomedin bioactivity in serum and amniotic fluid during pregnancy. J Clin Endocrinol Metab 1978;46:649. 464. Gautray JP, Jolivet A, Vielh JP, et al. Presence of immunoassayable beta-endorphin in human amniotic fluid: elevation in cases of fetal distress. Am J Obstet Gynecol 1977;129:211. 465. Hankins GD, Rowe J, Quirk JG, Jr., et al. Significance of brown and/or green amniotic fluid at the time of second trimester genetic amniocentesis. Obstet Gynecol 1984;64:353. 466. Szeto HH, Zervoudakis IA, Cederqvist LL, et al. Amniotic fluid transfer of meperidine from maternal plasma in early pregnancy. Obstet Gynecol 1978;52:59. 467. Broussard CS, Rasmussen SA, Reefhuis J, et al. Maternal treatment with opioid analgesics and risk for birth defects. Am J Obstet Gynecol 2011;204:314 e1. 468. Klotz U. The role of pharmacogenetics in the metabolism of antiepileptic drugs: pharmacokinetic and therapeutic implications. Clin Pharmacokinet 2007;46:271. 469. Jauniaux E, Gulbis B, Acharya G, et al. Maternal tobacco exposure and cotinine levels in fetal fluids in the first half of pregnancy. Obstet Gynecol 1999;93:25. 470. Barr DB, Bishop A, Needham LL. Concentration of xenobiotic chemicals in the maternal–fetal unit. Reprod Toxicol 2007;23:260. 471. Barr DB, Bishop A, Needham LL. Concentrations of xenobiotic chemicals in the maternal–fetal unit. Reprod Toxicol 2007;23:260.

472. Ostrea EM, Jr., Matias O, Keane C, et al. Spectrum of gestational exposure to illicit drugs and other xenobiotic agents in newborn infants by meconium analysis. J Pediatr 1998;133:513. 473. Bradman A, Barr DB, Claus Henn BG, et al. Measurement of pesticides and other toxicants in amniotic fluid as a potential biomarker of prenatal exposure: a validation study. Environ Health Perspect 2003;111: 1779. 474. Goh YI, Chudley AE, Clarren SK, et al. Development of Canadian screening tools for fetal alcohol spectrum disorder. Can J Clin Pharmacol 2008;15:e344. 475. Gray TR, Shakleya DM, Huestis MA. Quantification of nicotine, cotinine, trans-3′ -hydroxycotinine, nornicotine and norcotinine in human meconium by liquid chromatography/tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2008;863:107. 476. Ostrea EM, Morales V, Ngoumgna E, et al. Prevalence of fetal exposure to environmental toxins as determined by meconium analysis. Neurotoxicology 2002;23:329. 477. Custodio PJ, Carvalho ML, Nunes F, et al. Direct analysis of human blood (mothers and newborns) by energy dispersive X-ray fluorescence. J Trace Elem Med Biol 2005;19:151. 478. Turker G, Ergen K, Karakoc Y, et al. Concentrations of toxic metals and trace elements in the meconium of newborns from an industrial city. Biol Neonate 2006;89:244. 479. Schmidt D, Achermann J, Odermatt B, et al. Prenatally fabricated autologous human living heart valves based on amniotic fluid derived progenitor cells as single cell source. Circulation 2007;116:I64. 480. Sessarego N, Parodi A, Podesta M, et al. Multipotent mesenchymal stromal cells from amniotic fluid: solid perspectives for clinical application. Haematologica 2008;93:339. 481. Pozzobon M, Piccoli M, Schiavo AA, et al. Isolation of c-Kit+ human amniotic fluid stem cells from second trimester. Methods Mol Biol 2013;1035:191. 482. Vigeh M, Yokoyama K, Ramezanzadeh F, et al. Blood manganese concentrations and intrauterine growth restriction. Reprod Toxicol 2008;25:219. 483. Osada H, Watanabe Y, Nishimura Y, et al. Profile of trace element concentrations in the feto-placental unit in relation to fetal growth. Acta Obstet Gynecol Scand 2002;81:931. 484. Sakamoto M, Kubota M, Murata K, et al. Changes in mercury concentrations of segmental maternal hair during gestation and their correlations with other biomarkers of fetal exposure to methylmercury in the Japanese population. Environ Res 2008;106:270.

CHAPTER 3

Amniotic Fluid Constituents, Cell Culture, and Neural Tube Defects 189

485. Ozdemir HS, Karadas F, Pappas AC, et al. The selenium levels of mothers and their neonates using hair, breast milk, meconium, and maternal and umbilical cord blood in Van Basin. Biol Trace Elem Res 2008;122:206. 486. Tsutsumi O. Assessment of human contamination of estrogenic endocrine-disrupting chemicals and their risk for human reproduction. J Steroid Biochem Mol Biol 2005;93:325. 487. Willhite CC, Ball GL, McLellan CJ. Derivation of a bisphenol A oral reference dose (RfD) and drinking water equivalent concentration. J Toxicol Environ Health B Crit Rev 2008;11:69. 488. Ostrea EM, Jr., Bielawski DM, Posecion NC, Jr., et al. A comparison of infant hair, cord blood and meconium analysis to detect fetal exposure to environmental pesticides. Environ Res 2008;106:277. 489. Blount BC, Valentin-Blasini L. Biomonitoring as a method for assessing exposure to perchlorate. Thyroid 2007;17:837. 490. Wessels D, Barr DB, Mendola P. Use of biomarkers to indicate exposure of children to organophosphate pesticides: implications for a longitudinal study of children’s environmental health. Environ Health Perspect 2003;111:1939. 491. Whyatt RM, Barr DB, Camann DE, et al. Contemporary-use pesticides in personal air samples during pregnancy and blood samples at delivery among urban minority mothers and newborns. Environ Health Perspect 2003;111:749. 492. Jarrell J, Chan S, Hauser R, et al. Longitudinal assessment of PCBs and chlorinated pesticides in pregnant women from Western Canada. Environ Health 2005;4:10. 493. Kawashiro Y, Fukata H, Omori-Inoue M, et al. Perinatal exposure to brominated flame retardants and polychlorinated biphenyls in Japan. Endocr J 2008;55:1071. 494. Engel SM, Levy B, Liu Z, et al. Xenobiotic phenols in early pregnancy amniotic fluid. Reprod Toxicol 2006;21:110. 495. Mustafa AM, Malintan NT, Seelan S, et al. Phytoestrogens levels determination in the cord blood from Malaysia rural and urban populations. Toxicol Appl Pharmacol 2007;222:25. 496. Silva MJ, Reidy JA, Herbert AR, et al. Detection of phthalate metabolites in human amniotic fluid. Bull Environ Contam Toxicol 2004;72:1226. 497. Huang PC, Kuo PL, Chou YY, et al. Association between prenatal exposure to phthalates and the health of newborns. Environ Int 2009;35:14. 498. Milunsky A, Carmella SG, Ye M, et al. A tobacco-specific carcinogen in the fetus. Prenat Diagn 2000;20:307.

499. DeMarini DM. Genotoxicity of tobacco smoke and tobacco smoke condensate: a review. Mutat Res 2004;567:447. 500. Eyler FD, Behnke M, Wobie K, et al. Relative ability of biologic specimens and interviews to detect prenatal cocaine use. Neurotoxicol Teratol 2005;27:677. 501. Gray T, Huestis M. Bioanalytical procedures for monitoring in utero drug exposure. Anal Bioanal Chem 2007;388:1455. 502. Sandberg JA, Olsen GD. Microassay for the simultaneous determination of cocaine, norcocaine, benzoylecgonine and benzoylnorecgonine by high-performance liquid chromatography. J Chromatogr 1990;525:113. 503. Mastrogiannis DS, Decavalas GO, Verma U, et al. Perinatal outcome after recent cocaine usage. Obstet Gynecol 1990;76:8. 504. Abdeljaber M, Nolan BM, Schork MA. Maternal cocaine use during pregnancy: effect on the newborn infant. Pediatrics 1990;85:630. 505. Behnke M, Eyler FD, Garvan CW, et al. The search for congenital malformations in newborns with fetal cocaine exposure. Pediatrics 2001;107:E74. 506. Addis A,Moretti ME, Ahmed Syed F, et al. Fetal effects of cocaine: an updated meta-analysis. Reprod Toxicol 2001;15:341. 507. Gouin K, Murphy K, Shah PS. Effects of cocaine use during pregnancy on low birthweight and preterm birth: systematic review and metaanalyses. Am J Obstet Gynecol 2011;204:340 e1. 508. Hostetter A, Ritchie JC, Stowe ZN. Amniotic fluid and umbilical cord blood concentrations of antidepressants in three women. Biol Psychiatry 2000;48:1032. 509. Loughhead AM, Fisher AD, Newport DJ, et al. Antidepressants in amniotic fluid: another route of fetal exposure. Am J Psychiatry 2006;163:145. 510. Omtzigt JG, Nau H, Los FJ, et al. The disposition of valproate and its metabolites in the late first trimester and early second trimester of pregnancy in maternal serum, urine, and amniotic fluid: effect of dose, co-medication, and the presence of spina bifida. Eur J Clin Pharmacol 1992;43:381. 511. Lucas BR, Latimer J, Pinto RZ, et al. Gross motor deficits in children prenatally exposed to alcohol: a meta-analysis. Pediatrics 2014;134:e192. 512. Heller M, Burd L. Review of ethanol dispersion, distribution, and elimination from the fetal compartment. Birth Defects Res A Clin Mol Teratol 2014;100: 277. 513. Feeley M, Brouwer A. Health risks to infants from exposure to PCBs, PCDDs and PCDFs. Food Addit Contam 2000;17:325.

190 Genetic Disorders and the Fetus

514. Yamada H, Furuta I, Kato EH, et al. Maternal serum and amniotic fluid bisphenol A concentrations in the early second trimester. Reprod Toxicol 2002;16:735. 515. Ikezuki Y, Tsutsumi O, Takai Y, et al. Determination of bisphenol A concentrations in human biological fluids reveals significant early prenatal exposure. Hum Reprod 2002;17:2839. 516. Jedeon K, De la Dure-Molla M, Brookes SJ, et al. Enamel defects reflect perinatal exposure to bisphenol A. Am J Pathol 2013;183:108. 517. Zoeller RT, Bansal R, Parris C. Bisphenol-A, an environmental contaminant that acts as a thyroid hormone receptor antagonist in vitro, increases serum thyroxine, and alters RC3/neurogranin expression in the developing rat brain. Endocrinology 2005;146:607. 518. Whyatt RM, Barr DB. Measurement of organophosphate metabolites in postpartum meconium as a potential biomarker of prenatal exposure: a validation study. Environ Health Perspect 2001;109:417. 519. Fennell TR, Krol WL, Sumner SC, et al. Pharmacokinetics of dibutylphthalate in pregnant rats. Toxicol Sci 2004;82:407. 520. Cruger DG, Bruun-Petersen G, Kolvraa S. Early prenatal diagnosis: standard cytogenetic analysis of coelomic cells obtained by coelocentesis. Prenat Diagn 1996;16:945. 521. Elejalde BR, de Elejalde MM, Acuna JM, et al. Prospective study of amniocentesis performed between weeks 9 and 16 of gestation: its feasibility, risks, complications and use in early genetic prenatal diagnosis. Am J Med Genet 1990;35:188. 522. Daniel A, Ng A, Kuah KB, et al. A study of early amniocentesis for prenatal cytogenetic diagnosis. Prenat Diagn 1998;18:21. 523. Verma L, Macdonald F, Leedham P, et al. Rapid and simple prenatal DNA diagnosis of Down’s syndrome. Lancet 1998;352:9. 524. Pertl B, Kopp S, Kroisel PM, et al. Rapid detection of chromosome aneuploidies by quantitative fluorescence PCR: first application on 247 chorionic villus samples. J Med Genet 1999;36:300. 525. Lapidot-Lifson Y, Lebo RV, Flandermeyer RR, et al. Rapid aneuploid diagnosis of high-risk fetuses by fluorescence in situ hybridization. Am J Obstet Gynecol 1996;174:886. 526. D’Alton ME, Malone FD, Chelmow D, et al. Defining the role of fluorescence in situ hybridization on uncultured amniocytes for prenatal diagnosis of aneuploidies. Am J Obstet Gynecol 1997;176:769; discussion 74.

527. Kuo WL, Tenjin H, Segraves R, et al. Detection of aneuploidy involving chromosomes 13, 18, or 21, by fluorescence in situ hybridization (FISH) to interphase and metaphase amniocytes. Am J Hum Genet 1991; 49:112. 528. Ried T, Landes G, Dackowski W, et al. Multicolor fluorescence in situ hybridization for the simultaneous detection of probe sets for chromosomes 13, 18, 21, X and Y in uncultured amniotic fluid cells. Hum Mol Genet 1992;1:307. 529. Klinger K, Landes G, Shook D, et al. Rapid detection of chromosome aneuploidies in uncultured amniocytes by using fluorescence in situ hybridization (FISH). Am J Hum Genet 1992;51:55. 530. Ward BE, Gersen SL, Carelli MP, et al. Rapid prenatal diagnosis of chromosomal aneuploidies by fluorescence in situ hybridization: clinical experience with 4,500 specimens. Am J Hum Genet 1993;52:854. 531. Divane A, Carter NP, Spathas DH, et al. Rapid prenatal diagnosis of aneuploidy from uncultured amniotic fluid cells using five-colour fluorescence in situ hybridization. Prenat Diagn 1994;14:1061. 532. Gersen SL, Carelli MP, Klinger KW, et al. Rapid prenatal diagnosis of 14 cases of triploidy using fish with multiple probes. Prenat Diagn 1995;15:1. 533. Evans MI, Henry GP, Miller WA, et al. International, collaborative assessment of 146,000 prenatal karyotypes: expected limitations if only chromosome specific probes and fluorescent in-situ hybridization are used. Hum Reprod 1999;14:1213. 534. Feldman B, Ebrahim SA, Hazan SL, et al. Routine prenatal diagnosis of aneuploidy by FISH studies in high-risk pregnancies. Am J Med Genet 2000;90:233. 535. Oneda B, Baldinger R, Reissmann R, et al. High-resolution chromosomal microarrays in prenatal diagnosis significantly increase diagnostic power. Prenat Diagn 2014;34:525. 536. Committee Opinion No. 581: the use of chromosomal microarray analysis in prenatal diagnosis. Obstet Gynecol 2013;122:1374. 537. Brady PD, Delle Chiaie B, Christenhusz G, et al. A prospective study of the clinical utility of prenatal chromosomal microarray analysis in fetuses with ultrasound abnormalities and an exploration of a framework for reporting unclassified variants and risk factors. Genet Med 2014;16:469. 538. Donnenfeld AE, Lockwood D, Custer T, et al. Prenatal diagnosis from fetal urine in bladder outlet obstruction: success rates for traditional cytogenetic evaluation and interphase fluorescence in situ hybridization. Genet Med 2002;4:444. 539. Ville YG, Nicolaides KH, Campbell S. Prenatal diagnosis of fetal malformations by ultrasound. In: Milunsky

CHAPTER 3

Amniotic Fluid Constituents, Cell Culture, and Neural Tube Defects 191

A, ed. Genetic disorders and the fetus: diagnosis, prevention, and treatment. Baltimore, MD: The Johns Hopkins University Press, 2004:836. 540. Golden WL, Schneider BF, Gustashaw KM, et al.

553.

Prenatal diagnosis of Turner syndrome using cells cultured from cystic hygromas in two pregnancies with normal maternal serum alpha-fetoprotein. Prenat

554.

Diagn 1989;9:683. 541. Wax JR, Blakemore KJ, Soloski MJ, et al. Fetal ascitic fluid: a new source of lymphocytes for rapid chromo-

555.

somal analysis. Obstet Gynecol 1992;80:533. 542. Costa D, Borrell A, Margarit E, et al. Rapid fetal karyotype from cystic hygroma and pleural effusions. Pre-

556.

nat Diagn 1995;15:141. 543. Wald NJ, Rodeck C, Hackshaw AK, et al. First and second trimester antenatal screening for Down’s syndrome: the results of the Serum, Urine and Ultrasound

557.

Screening Study (SURUSS). Health Technol Assess 2003;7:1. 544. van Dyke DL, Ebrahim SA, Al Saadi AA, et al. The impact of maternal serum screening programs for

558.

Down syndrome in southeast Michigan, 1988–2003. Prenat Diagn 2007;27:583. 545. Bianchi DW, Parker RL, Wentworth J, et al. DNA sequencing versus standard prenatal aneuploidy

559.

screening. N Engl J Med 2014;370:799. 546. Grati FR, Malvestiti F, Ferreira JC, et al. Fetoplacental mosaicism: potential implications for false-positive and false-negative noninvasive prenatal screening

560.

results. Genet Med 2014;16:620. 547. Wang JC, Sahoo T, Schonberg S, et al. Discordant noninvasive prenatal testing and cytogenetic results: a

561.

study of 109 consecutive cases. Genet Med 2014. 548. Holbrook KA, Odland GF. The fine structure of developing human epidermis: light, scanning, and transmis-

562.

sion electron microscopy of the periderm. J Invest Dermatol 1975;65:16. 549. Holbrook KA, Odland GF. Regional development of

563.

the human epidermis in the first trimester embryo and the second trimester fetus (ages related to the timing

564.

of amniocentesis and fetal biopsy). J Invest Dermatol 1980;74:161. 550. Tyden O, Bergstrom S, Nilsson BA. Origin of amniotic

565.

fluid cells in mid-trimester pregnancies. Br J Obstet Gynaecol 1981;88:278. 551. Gosden CM, Brock DJ. Morphology of rapidly adhering amniotic-fluid cells as an aid to the diagnosis of neural tube defects. Lancet 1977;i:919. 552. Huisjes HJ. Cytology of the amniotic fluid and its clinical applications. In: Fairweather DVI, Eskes TKAB,

566.

eds. Amniotic fluid research and clinical applications, 2nd edn. Amsterdam: Elsevier/North Holland, 1978:93. Bergström S. Ultrastructure of cell detachment from the human fetus in early pregnancy. Acta Obstet Gynecol Scand 1979;58:353. Schrage R, Bogelspacher HR, Wurster KG. Amniotic fluid cells in the second trimester of pregnancy. Acta Cytol 1982;26:407. Agorastos T, Bar T, Grussendorf EI, et al. Ultrastructural aspects of amniotic-fluid cells: B. Vital cells. Z Geburtshilfe Perinatol 1982;186:41. Hoehn H, Bryant EM, Karp LE, et al. Cultivated cells from diagnostic amniocentesis in second trimester pregnancies. I. Clonal morphology and growth potential. Pediatr Res 1974;8:746. Felix JS, Doherty RA, Davis HT, et al. Amniotic fluid cell culture. I. Experimental design for evaluating cell culture variables: determination of optimal fetal calf serum concentration. Pediatr Res 1974;8:870. Polgar K, Adany R, Abel G, et al. Characterization of rapidly adhering amniotic fluid cells by combined immunofluorescence and phagocytosis assays. Am J Hum Genet 1989;45:786. Greenebaum E, Mansukhani MM, Heller DS, et al. Open neural tube defects: immunocytochemical demonstration of neuroepithelial cells in amniotic fluid. Diagn Cytopathol 1997;16:143. Medina-Gomez P, McBride WH. Amniotic fluid macrophages fromnormal and malformed fetuses. Prenat Diagn 1986;6:195. Roberts SH, Little E, Vaughan M, et al. Rapid prenatal diagnosis of Patau’s syndrome in a fetus with an abdominal wall defect by 72 hour culture of cells from amniotic fluid. Prenat Diagn 1993;13:971. Gosden C, Brock DJ. Amniotic fluid cell morphology in early antenatal prediction of abortion and low birth weight. Br Med J 1978;2:1186. Gosden CM. Amniotic fluid cell types and culture. Br Med Bull 1983;39:348. Osborn M, Weber K. Tumor diagnosis by intermediate filament typing: a novel tool for surgical pathology. Lab Invest 1983;48:372. Franke WW, Appelhans B, Schmid E, et al. Identification and characterization of epithelial cells in mammalian tissues by immunofluorescence microscopy using antibodies to prekeratin. Differentiation 1979; 15:7. Priest RE, Priest JH, Laundon CH, et al. Multinucleate cells incultures of human amniotic fluid form by fusion. Lab Invest 1980;43:140.

192 Genetic Disorders and the Fetus

567. Medina-Gomez P, Johnston TH. Cell morphology in long-term cultures of normal and abnormal amniotic fluids. Hum Genet 1982;60:310. 568. Megaw JM, Priest JH, Priest RE, et al. Differentiation in human amniotic fluid cell cultures: II: Secretion of an epithelial basement membrane glycoprotein. J Med Genet 1977;14:163. 569. Priest RE, Priest JH, Moinuddin JF, et al. Differentiation in human amniotic fluid cell cultures: I: Collagen production. J Med Genet 1977;14:157. 570. Priest RE, Marimuthu KM, Priest JH. Origin of cells in human amniotic fluid cultures: ultrastructural features. Lab Invest 1978;39:106. 571. Priest RE, Priest JH, Moinuddin JF, et al. Differentiation in human amniotic fluid cell cultures: chorionic gonadotropin production. In Vitro 1979;15:142. 572. Thakar N, Priest RE, Priest JH. Estrogen production by cultured amniotic fluid cells. Clin Res 1982;30:888A. 573. O’Shannessy DJ, Priest RE, Priest JH. Metabolism of [4-14C]androstenedione by cells cultured from human amniotic fluid. J Steroid Biochem 1984;20:935. 574. Laundon CH, Priest JH, Priest RE. The characterization of hCG regulation in cultured human amniotic fluid cells. Prenat Diagn 1981;1:269. 575. Laundon CH, Priest JH, Priest RE. Characterization of hCG regulation in cultured human amniotic fluid cells: II. Mechanisms for stimulation. In Vitro 1983;19:911. 576. Chang HC, Jones OW. In vitro characteristics of human fetal cells obtained from chorionic villus sampling and amniocentesis. Prenat Diagn 1988;8:367. 577. Whitsett CF, Priest JH, Priest RE, et al. HLA typing of cultured amniotic fluid cells. Am J Clin Pathol 1983;79:186. 578. Crouch E, Bornstein P. Collagen synthesis by human amniotic fluid cells in culture: characterization of a procollagen with three identical proalpha1(I) chains. Biochemistry 1978;17:5499. 579. Crouch E, Bornstein P. Characterization of a type IV procollagen synthesized by human amniotic fluid cells in culture. J Biol Chem 1979;254:4197. 580. Crouch E, Balian G, Holbrook K, et al. Amniotic fluid fibronectin. Characterization and synthesis by cells in culture. J Cell Biol 1978;78:701. 581. Bryant EM, Crouch E, Bornstein P, et al. Regulation of growth and gene activity in euploid hybrids between human neonatal fibroblasts and epithelioid amniotic fluid cells. Am J Hum Genet 1978;30:392. 582. Johnston P, Salk D, Martin GM, et al. Cultivated cells from mid-trimester amniotic fluids: IV. Cell type identification via one and two-dimensional electrophoresis of clonal whole cell homogenates. Prenat Diagn 1982;2:79.

583. Virtanen I, von Koskull H, Lehto VP, et al. Cultured human amniotic fluid cells characterized with antibodies against intermediate filaments in indirect immunofluorescence microscopy. J Clin Invest 1981; 68:1348. 584. Cremer M, Treiss I, Cremer T, et al. Characterization of cells of amniotic fluids by immunological identification of intermediate-sized filaments: presence of cells of different tissue origin. Hum Genet 1981;59:373. 585. Chen WW. Studies on the origin of human amniotic fluid cells by immunofluorescent staining of keratin filaments. J Med Genet 1982;19:433. 586. Moll R, Franke WW, Schiller DL, et al. The catalog of human cytokeratins: patterns of expression in normal epithelia, tumors and cultured cells. Cell 1982; 31:11. 587. Ochs BA, Franke WW, Moll R, et al. Epithelial character and morphologic diversity of cell cultures from human amniotic fluids examined by immunofluorescence microscopy and gel electrophoresis of cytoskeletal proteins. Differentiation 1983;24:153. 588. Hoehn H, Bryant EM, Karp LE, et al. Cultivated cells from diagnostic amniocentesis in second trimester pregnancies. II. Cytogenetic parameters as functions of clonal type and preparative technique. Clin Genet 1975;7:29. 589. Regauer S, Franke WW, Virtanen I. Intermediate filament cytoskeleton of amnion epithelium and cultured amnion epithelial cells: expression of epidermal cytokeratins in cells of a simple epithelium. J Cell Biol 1985;100:997. 590. Hsu LY, Kaffe S, Perlis TE. A revisit of trisomy 20 mosaicism in prenatal diagnosis – an overview of 103 cases. Prenat Diagn 1991;11:7. 591. Van Dyke DL, Roberson JR, Babu VR, et al. Trisomy 20 mosaicism identified prenatally and confirmed in foreskin fibroblasts. Prenat Diagn 1989;9:601. 592. Bell JE, Barron L, Raab G. Antenatal detection of neural tube defects: comparison of biochemical and immunofluorescence methods. Prenat Diagn 1994;14:615. 593. Melancon SB, Lee SY, Nadler HL. Histidase activity in cultivated human amniotic fluid cells. Science 1971;173:627. 594. Gerbie AB, Melancon SB, Ryan C, et al. Cultivated epithelial-like cells and fibroblasts from amniotic fluid: their relationship to enzymatic and cytologic analysis. Am J Obstet Gynecol 1972;114:314. 595. Van der Veer E, Kleijer WJ, de Josselin de Jong JE, Galjaard H. Lysosomal enzyme activities in different types of amniotic fluid cells measured by microchemical methods, combined with interference microscopy. Hum Genet 1978;40:285.

CHAPTER 3

Amniotic Fluid Constituents, Cell Culture, and Neural Tube Defects 193

596. Burton BK, Gerbie AB, Nadler HL. Biochemical and biological problems and pitfalls of cell culture for prenatal diagnosis. In: Milunsky A, ed. Genetic disorders and the fetus: diagnosis prevention, and treatment. New York: Plenum Press, 1979:369. 597. Hoehn H, Bryant EM, Fantel AG, et al. Cultivated cells from diagnostic amniocentesis in second trimester pregnancies. III. The fetal urine as a potential source of clonable cells. Humangenetik 1975;29:285. 598. Chang HC, Jones OW. Amniocentesis: cell culture of human amniotic fluid in a hormone supplement. In: Sirbasku DA, Pardee AB, Sato GH, eds. Growth of cells in hormonally defined media (Cold Spring Harbor Conferences on Cell Proliferation). Cold Spring Harbor, NY: Cold Spring Harbor Laboratory Press, 1982:1187. 599. Medina-Gomez P, Bard JB. Analysis of normal and abnormal amniotic fluid cells in vitro by cinemicrography. Prenat Diagn 1983;3:311. 600. Harris A. Glycoproteins that distinguish different cell types found in amniotic fluid. Hum Genet 1982;62:188. 601. Felix JS, Sun TT, Littlefield JW. Human epithelial cells cultured from urine: growth properties and keratin staining. In Vitro 1980;16:866. 602. von Koskull H, Aula P, Trejdosiewicz LK, et al. Identification of cells from fetal bladder epithelium in human amniotic fluid. Hum Genet 1984;65:262. 603. Richkind KE, Risch NJ. Sensitivity of chromosomal mosaicism detection by different tissue culture methods. Prenat Diagn 1990;10:519. 604. Hoehn HW. Fluid cell culture. In: Milunsky A, ed. Genetic disorders and the fetus, 4th edn. Baltimore, MD: The Johns Hopkins University Press, 1998:128. 605. Kennerknecht I, Baur-Aubele S, Grab D, et al. First trimester amniocentesis between the seventh and 13th weeks: evaluation of the earliest possible genetic diagnosis. Prenat Diagn 1992;12:595. 606. Casey ML, Word RA, MacDonald PC. Endothelin-1 gene expression and regulation of endothelin mRNA and protein biosynthesis in avascular human amnion. Potential source of amniotic fluid endothelin. J Biol Chem 1991;266:5762. 607. Chang HC, Jones OW. Reduction of sera requirements in amniotic fluid cell culture. Prenat Diagn 1985;5:305. 608. Liu L, Brinkman A, Blat C, et al. IGFBP-1, an insulin like growth factor binding protein, is a cell growth inhibitor. Biochem Biophys Res Commun 1991;174:673. 609. Priest JH, Rao KW. Prenatal chromosome diagnosis. In: Barch MJ, ed. The AGT cytogenetics laboratory manual, 3rd edn. Philadelphia: Lippincott Williams & Wilkins, 1997:199.

610. Schmid W. A technique for in situ karyotyping of primary amniotic fluid cell cultures. Humangenetik 1975;30:325. 611. Hoehn H, Rodriguez ML, Norwood TH, et al. Mosaicism in amniotic fluid cell cultures: classification and significance. Am J Med Genet 1978;2:253. 612. Boué J, Nicolas H, Barichard F, et al. Le clonage des cellules du liquide amniotique, aide dans l’interpretation des mosaiques chromosomiques en diagnostic prenatal. Ann Genet 1979;22:3. 613. Hecht F, Peakman DC, Kaiser-McCaw B, et al. Amniocyte clones for prenatal cytogenetics. Am J Med Genet 1981;10:51. 614. Tabor A, Lind AM, Andersen AM, et al. A culture vessel for amniotic fluid cells allowing faster preparation of chromosome slides. Prenat Diagn 1984;4:451. 615. Benn PA, Hsu LY. Maternal cell contamination of amniotic fluid cell cultures: results of a U.S. nationwide survey. Am J Med Genet 1983;15:297. 616. Bui TH, Iselius L, Lindsten J. European collaborative study on prenatal diagnosis: mosaicism, pseudomosaicism and single abnormal cells in amniotic fluid cell cultures. Prenat Diagn 1984;4 Spec No:145. 617. Worton RG, Stern R. A Canadian collaborative study of mosaicism in amniotic fluid cell cultures. Prenat Diagn 1984;4 Spec No:131. 618. Batanian JR, Ledbetter DH, Fenwick RG. A simple VNTR-PCR method for detecting maternal cell contamination in prenatal diagnosis. Genet Test 1998;2:347. 619. Frederickson RM, Wang HS, Surh LC. Some caveats in PCR-based prenatal diagnosis on direct amniotic fluid versus cultured amniocytes. Prenat Diagn 1999;19:113. 620. Smith GW, Graham CA, Nevin J, et al. Detection of maternal cell contamination in amniotic fluid cell cultures using fluorescent labelled microsatellites. J Med Genet 1995;32:61. 621. Claussen U, Schafer H, Trampisch HJ. Exclusion of chromosomal mosaicism in prenatal diagnosis. Hum Genet 1984;67:23. 622. Sikkema-Raddatz B, Castedo S, TeMeerman GJ. Probability tables for exclusion of mosaicism in prenatal diagnosis. Prenat Diagn 1997;17:115. 623. American College of Medical Genetics. Standards and guidelines for clinical genetics laboratories, 2nd edn. Bethesda, MD: American College of Medical Genetics, 1999. 624. American College of Medical Genetics. Standards and guidelines for clinical genetics laboratories, 2008. https://www.acmg.net/ACMG/Medical-GeneticsPractice-Resources/Technical_Standards_and_ Guidelines.aspx (accessed July 8, 2020).

194 Genetic Disorders and the Fetus

625. Spurbeck JL, Zinsmeister AR, Meyer KJ, et al. Dynamics of chromosome spreading. Am J Med Genet 1996;61:387. 626. Henegariu O, Heerema NA, Lowe Wright L, et al. Improvements in cytogenetic slide preparation: controlled chromosome spreading, chemical aging and gradual denaturing. Cytometry 2001;43:101. 627. Spurbeck JL. The dynamics of chromosome spreading. Compact disk copyright 2013. Distributed by Association of Genetic Technologists (AGT), 2014. 628. Cheung SW, Spitznagel E, Featherstone T, et al. Exclusion of chromosomal mosaicism in amniotic fluid cultures: efficacy of in situ versus flask techniques. Prenat Diagn 1990;10:41. 629. Featherstone T, Cheung SW, Spitznagel E, et al. Exclusion of chromosomal mosaicism in amniotic fluid cultures: determination of number of colonies needed for accurate analysis. Prenat Diagn 1994;14:1009. 630. Cheng EY, Luthy DA, Dunne DF, et al. Is the 15-in situ clone protocol necessary to detect amniotic fluid mosaicism? Am J Obstet Gynecol 1995;173:1025. 631. Ing PS, van Dyke DL, Caudill SP, et al. Detection of mosaicism in amniotic fluid cultures: a CYTO2000 collaborative study. Genet Med 1999;1:94. 632. American College of Medical Genetics. Standards and guidelines for clinical genetics laboratories. E4.1.3 amniotic fluid. Bethesda, MD: American College of Medical Genetics, 2006. 633. Miron PM. Preparation, culture, and analysis of amniotic fluid samples. Curr Protoc Hum Genet 2012:8.4. 634. van Dyke DL, Roberson JR,Wiktor A. Prenatal cytogenetic diagnosis. In: McClatchey KD, ed. Clinical laboratory medicine, 2nd edn. Philadelphia: Lippincott Williams & Wilkins, 2002:636. 635. Spurbeck JL, Carlson RO, Allen JE, et al. Culturing and robotic harvesting of bone marrow, lymph nodes, peripheral blood, fibroblasts, and solid tumors with in situ techniques. Cancer Genet Cytogenet 1988;32:59. 636. Carlan SJ, Papenhausen P, O’Brien WF, et al. Effect of maternal–fetal movement on concentration of cells obtained at genetic amniocentesis. Am J Obstet Gynecol 1990;163:490. 637. Fischer RL, LaMotta J, McMorrow LE, et al. Effect of pre-amniocentesis uterine manipulation on amniocyte concentration and culture duration: a randomized, clinical trial. Prenat Diagn 1996;16:673. 638. Melnyk JH, Persinger G, Teplitz RLA. A micromethod for processing amniotic fluid cells. In Vitro 1979;15:200. 639. Byrne DL, Marks K, Braude PR, et al. Amniofiltration in the first trimester: feasibility, technical aspects

640.

641.

642.

643.

644.

645.

646.

647.

648. 649.

650.

651.

652. 653.

654.

and cytological outcome. Ultrasound Obstet Gynecol 1991;1:320. Sundberg K, Smidt-Jensen S, Lundsteen C, et al. Filtration and recirculation of early amniotic fluid. Evaluation of cell cultures from 100 diagnostic cases. Prenat Diagn 1993;13:1101. Sundberg K, Bang J, Brocks V, et al. Early sonographically guided amniocenteses with filtration technique: follow-up on 249 procedures. J Ultrasound Med 1995;14:585. Sundberg K, Lundsteen C, Philip J. Early filtration amniocentesis for further investigation of mosaicism diagnosed by chorionic villus sampling. Prenat Diagn 1996;16:1121. Kunsela P, Seppala MB, Brock DJH, et al. Amniotic fluid fibronectin in normal pregnancy and in pregnancies with anencephalic fetus. Biomedicine 1978;29:296. Vlodavsky I, Voss R, Yarkoni S, et al. Stimulation of human amniotic fluid cell proliferation and colony formation by cell plating on a naturally produced extracellular matrix. Prenat Diagn 1982;2:13. Crickard K, Golbus MS. Influence of extracellular matrix on the proliferation of human amniotic fluid cells in vitro. Prenat Diagn 1982;2:89. Brackertz M, Kubbies M, Feige A, et al. Decreased oxygen supply enhances growth in culture of human midtrimester amniotic fluid cells. Hum Genet 1983;64:334. Held KR, Sönnichsen S. The effect of oxygen tension on colony formation and cell proliferation of amniotic fluid cells in vitro. Prenat Diagn 1984;4:171. Barnes D, Sato G. Serum-free cell culture: a unifying approach. Cell 1980;22:649. Gospodarowicz D, Moran JS, Owashi ND. Effects of fibroblast growth factor and epidermal growth factor on the rate of growth of amniotic fluid-derived cells. J Clin Endocrinol Metab 1977;44:651. Porreco RP, Bradshaw C, Sarkar S, et al. Enhanced growth of amniotic fluid cells in presence of fibroblast growth factor. Obstet Gynecol 1980;55:55. Art to Science. Freezing and thawing serum and other biological materials: optimal procedures minimize damage and maximize shelf-life. HyClone Laboratories, Inc, 1992;11:1. Chang HC, Jones OW. A new growth medium for human amniotic fluid cells. Karyogram 1981;7:54. Chang HC, Jones OW, Masui H. Human amniotic fluid cells grown in a hormone-supplemented medium: suitability for prenatal diagnosis. Proc Natl Acad Sci U S A 1982;79:4795. Biddle WC, Kuligowski S, Lockwood DH, et al. AmnioMAXTM-C100: a new specialized cell culture

CHAPTER 3

655.

656.

657.

658.

659.

660.

661.

662.

663. 664.

665.

666. 667. 668. 669.

670.

Amniotic Fluid Constituents, Cell Culture, and Neural Tube Defects 195

medium for the propagation of human amniocytes. Focus 1992;14:80. Claussen U. The pipette method: a new rapid technique for chromosome analysis in prenatal diagnosis. Hum Genet 1980;54:277. Salk D, Disteche C, Stenchever MR, et al. Routine use of Chang medium for prenatal diagnosis: improved growth and increased chromosomal breakage. Am J Hum Genet 1983;35:151A. Masia A, Jenkins EC, Duncan C. Chromosomal abnormalities in cultures with Chang medium. Am J Hum Genet 1986;39:260. Krawczun MS, Jenkins EC, Masia A, et al. Chromosomal abnormalities in amniotic fluid cell cultures: a comparison of apparent pseudomosaicism in Chang and RPMI-1640 media. Clin Genet 1989;35:139. Eiben B, Goebel R, Hansen S, et al. Early amniocentesis – a cytogenetic evaluation of over 1500 cases. Prenat Diagn 1994;14:497. Assel BG, Lewis SM, Dickerman LH, et al. Single operator comparison of early and mid-second-trimester amniocentesis. Obstet Gynecol 1992;79:940. Corning guide for identifying and correcting common cell growth problems. http://www.bioprocessonline. com/doc/identifying-correcting-cell-growthproblems0001 (accessed May 19, 2015). Persutte WH, Lenke RR. Failure of amniotic-fluid cell growth: is it related to fetal aneuploidy? Lancet 1995;345:96. Elejalde BR, de Elejalde MM, Soto A. Amniotic-fluid cell growth and fetal aneuploidy. Lancet 1995;345:924. Lam YH, Tang MH, Sin SY, et al. Clinical significance of amniotic-fluid-cell culture failure. Prenat Diagn 1998;18:343. American College of Medical Genetics Standards and Guidelines for Clinical Genetics Laboratories. https:// www.acmg.net/ACMG/Medical-Genetics-PracticeResources/Technical_Standards_and_Guidelines.aspx (accessed July 8, 2020). Milunsky A, Bender CS. Failure of amniotic-fluid cell growth with toxic tubes. N Engl J Med 1979;301:47. Kohn G. Failure of amniotic fluid cell cultures due to syringe toxicity. Prenat Diagn 1981;1:233. Chiesa J, Bureau JP. Nothing ventured, nothing gained! Prenat Diagn 1999;19:894. Seguin LR, Palmer CG. Variables influencing growth and morphology of colonies of cells from human amniotic fluid. Prenat Diagn 1983;3:107. Felix JS, Doherty RA. Amniotic fluid cell culture II. Evaluation of a red blood cell lysis procedure for culture of cells from blood-contaminated amniotic fluid. Clin Genet 1979;15:215.

671. Johansson KE, Bolske G. Evaluation and practical aspects of the use of a commercial DNA probe for detection of mycoplasma infections in cell cultures. J Biochem Biophys Methods 1989;19:185. 672. Knutsen T. Laboratory safety, quality control and regulations. In: Barch MJ, ed. The AGT cytogenetics laboratory manual, 3rd edn. Philadelphia: Lippincott Williams & Wilkins, 1997:597. 673. Holtge GA. Laboratory safety. In: McClatchey KD, ed. Clinical laboratory medicine, 2nd edn. Philadelphia: Lippincott Williams & Wilkins, 2002:78. 674. Travers EM. Basic laboratory management. In: McClatchey KD, ed. Clinical laboratory medicine, 2nd edn. Philadelphia: Lippincott Williams & Wilkins, 2002:3. 675. Tsai MS, Lee JL, Chang YJ, et al. Isolation of human multipotent mesenchymal stem cells from second-trimester amniotic fluid using a novel two-stage culture protocol. Hum Reprod 2004;19:1450. 676. De Coppi P, Bartsch G, Jr., Siddiqui MM, et al. Isolation of amniotic stem cell lines with potential for therapy. Nat Biotechnol 2007;25:100. 677. Guillot PV, Gotherstrom C, Chan J, et al. Human first trimester fetal MSC express pluripotency markers and grow faster and have longer telomeres than adult MSC. Stem Cells 2007;25:646. 678. Roubelakis MG, Pappa KI, Bitsika V, et al. Molecular and proteomic characterization of human mesenchymal stem cells derived from amniotic fluid: comparison to bone marrow mesenchymal stem cells. Stem Cells Dev 2007;16:931. 679. Zhou J,Wang D, Liang T, et al. Amniotic fluid-derived mesenchymal stem cells: characteristics and therapeutic applications. Arch Gynecol Obstet 2014;290:223. 680. In’t Anker PS, Scherjon SA, Kleijburg-van der Keur C, et al. Amniotic fluid as a novel source of mesenchymal stem cells for therapeutic transplantation. Blood 2003;102:1548. 681. Trounson A. A fluid means of stem cell generation. Nat Biotechnol 2007;25:62. 682. Liu YW, Roan JN, Wang SP, et al. Xenografted human amniotic fluid-derived stem cell as a cell source in therapeutic angiogenesis. Int J Cardiol 2013; 168:66. 683. Petsche Connell J, Camci-Unal G, Khademhosseini A, et al. Amniotic fluid-derived stem cells for cardiovascular tissue engineering applications. Tissue Eng Part B Rev 2013;19:368. 684. Kang NH, Hwang KA, Kim SU, et al. Potential antitumor therapeutic strategies of human amniotic membrane and amniotic fluid-derived stem cells. Cancer Gene Ther 2012;19:517.

196 Genetic Disorders and the Fetus

685. Kaviani A, Perry TE, Dzakovic A, et al. The amniotic fluid as a source of cells for fetal tissue engineering. J Pediatr Surg 2001;36:1662. 686. Gucciardo L, Lories R, Ochsenbein-Kolble N, et al. Fetal mesenchymal stem cells: isolation, properties

and potential use in perinatology and regenerative medicine. BJOG 2009;116:166. 687. Murphy SV, Atala A. Amniotic fluid and placental membranes: unexpected sources of highly multipotent cells. Semin Reprod Med 2013;31:62.

4

Molecular Aspects of Placental Development Wendy P. Robinson 1,2 and Deborah E. McFadden 1,2 1 2

BC Children’s Hospital Research Institute, Vancouver, BC, Canada University of British Columbia, Vancouver, BC, Canada

Overview The placenta is a fetal organ that is discarded after birth, but is essential to ensuring normal development in utero. It regulates fetal growth, protects the fetus from infection and other adverse exposures, as well as generally programming the fetus for good health after birth. Screening for placental disease is an important component to the assessment of the fetus in pregnancy. Reduced placental efficiency can lead to fetal growth restriction (FGR) and/or maternal preeclampsia (PE). This can be caused by genetic changes within the placenta or by environmental influences, such as maternal stress or drug exposure. In this chapter, causes of placental disease and the role of the placenta in diagnosis of fetal health will be reviewed with a focus on genetic associations.

Placental structure Evaluating the placenta requires an understanding of its unique structure and development. The chorionic villi that compose the placenta are organized into 50–70 distinct tree-like structures that grow in a clonal manner outwards from the chorionic plate into the basal plate (which interface with the maternal decidua).1 These villi are bathed in maternal blood, from which they sponge up nutrients important for fetal growth.

The maternal blood is in direct contact with the outer trophoblast bilayer of the chorionic villi. This bilayer is made up of a multinucleated syncytium derived by fusion of the cytotrophoblast cells that form a single-cell layer below the syncytium. In addition, some cytotrophoblasts form columns that migrate into and anchor the placenta to the uterine wall. Invasive cells that detach from these columns are termed extravillous trophoblasts (EVTs) and include the interstitial cytotrophoblasts (iCTBs) found in the decidual stroma and those that remodel maternal blood vessels, termed endovascular cytotrophoblasts (eCTBs).1 The inner core of the villi is the chorionic mesenchyme, which includes structural components, and a mix of cells including fetal blood vessels, fibroblasts, pericytes, and Hofbauer cells (placental macrophages). These extraembryonic cells derive from the epiblast of the blastocyst, from which the fetus is also derived. Placental size is strongly correlated with fetal size; however, there is considerable variation in placental size for any given birthweight.2 The efficiency of the placenta depends on the surface area for exchange, thickness, and density of transporter proteins,3 and birthweight is highly associated with placental weight.4 Interestingly, mean placental size can vary between populations and even within a population over time because of changes in maternal nutrition or other environmental conditions.5

Genetic Disorders and the Fetus: Diagnosis, Prevention, and Treatment, Eighth Edition. Edited by Aubrey Milunsky and Jeff M. Milunsky. © 2021 Aubrey Milunsky and Jeff M. Milunsky. Published 2021 by John Wiley & Sons Ltd.

197

198 Genetic Disorders and the Fetus

Placental development and function The placenta is responsible for many functions, which change as pregnancy proceeds.1 In early gestation, the primary roles of the placenta include invasion into the maternal endometrium, remodeling of maternal vasculature, and secretion of hormones important to maintain pregnancy. The placenta subsequently regulates blood flow and nutrient delivery to the fetus, buffers the fetus from adverse environmental effects, and generally performs the functions of multiple organs (lung, brain, kidney, immune system, etc.). Implantation

During the invasion process, the early trophoblasts produce molecules to help them attach to and invade the uterine wall (e.g. integrins), prevent menstruation (e.g. human chorionic gonadotropin (hCG)), destroy the uterine matrix (e.g. matrix metalloproteinases), and suppress the maternal immune system (e.g. corticotropin-releasing hormone (CRH)).6, 7 hCG (encoded by CGA and CGB) is one of the earliest hormones expressed from syncytiotrophoblast and stimulates many other processes. Multiple growth factors are important in regulating trophoblast proliferation, including placental growth factor (PlGF), epidermal growth factor (EGF), and transforming growth factor β (TGF-β). In addition, microparticles, such as microvesicles (0.1–2 μm) and exosomes (30–100 nm), are released by the placental syncytium into maternal decidua and blood and may play a role in early maternal immune suppression and vascular remodeling.8–11 These microparticles provide a rich source of placental derived material detectable in maternal blood early in pregnancy that has the potential to be used in monitoring the pregnancy. Failure of implantation and invasion can lead to early miscarriage. The majority of miscarriages occurring in the first trimester of pregnancy are associated with chromosome abnormalities, with trisomy, triploidy, and 45,X accounting for the vast majority of these.12, 13 The reasons for implantation failure in chromosomally abnormal cases are likely complex, involving dysregulation of multiple important genes that then impede trophoblast growth and invasion. Trisomy for chromosomes

1, 11, and 19 are rarely observed even in early miscarriages, and presumably do not survive to clinical detection of pregnancy. Chromosome 19 not only has the highest gene density of any chromosome, but is sometimes referred to as the “placenta chromosome” because of the numerous placenta-specific genes located on it, including the highly expressed pregnancy-specific glycoprotein cluster (PSG),14 and the maternally imprinted chromosome 19 miRNA cluster (C19MC), the largest microRNA cluster in humans.15 Failure of implantation among genetically normal conceptuses can be due to a nonreceptive maternal environment resulting from disturbances in maternal hormone levels, immune health, anatomical interference, or a variety of maternal health conditions.16 Angiogenesis

Placental angiogenesis and vasculogenesis serves to increase both uterine (maternal) and umbilical (fetal) blood flow. This process is dependent on a balance between pro- and antiangiogenic factors.17 Early in pregnancy, EVTs invade and plug the maternal uterine arteries, helping to maintain a low-oxygen environment needed for trophoblast proliferation.18 Trophoblast cells (eCTBs) also migrate along the lumina of spiral arterioles, replacing the maternal endothelial lining. This expands the diameter of the maternal vessels and, in combination with gradual disintegration of the spiral artery plugs, results in a dramatic increase in blood flow after 12 weeks gestational age, which is needed to support fetal growth later in pregnancy. In turn, placental vasculature develops, and increases throughout gestation as the needs of the fetus grow.19 FGR can result from poor spiral artery remodeling or reduced vascular development within the placenta. Furthermore, insufficient remodeling of the maternal spiral arteries can result in a prolonged state of hypoxia and increased reoxygenation stress. This leads to increased syncytiotrophoblast apoptosis and necrosis, causing increased debris circulating in the maternal blood that has been associated with maternal PE.20 In addition to FGR and PE, abnormal spiral artery remodeling has been associated with placental abruption, preterm premature rupture of membranes, and intrauterine fetal death.21

CHAPTER 4

Nutrient delivery

Fetal growth is dependent on efficient nutrient delivery to the fetus. This is determined by maternal availability, maternal blood flow to the placenta, the amount of placental surface in contact with maternal blood, and the efficiency of placental transport.22, 23 Transport of substances across the placenta can occur by (i) passive transport (simple or facilitated diffusion); (ii) active transport; and (iii) vesicular transport, by which large molecules are captured by microvesicles. A well-functioning placenta can be extremely efficient at extracting nutrients for the fetus even when maternal supplies are low. For example, there is a threefold increase in folate concentration in the placenta compared with maternal blood;24 this is accomplished via several folate receptors highly expressed in the human placenta, including folate receptor 1 (FOLR1), proton-coupled high-affinity folate transporter (PCFT), and reduced folate carrier (RFC).25 As the fetus grows and requires more nutrients, the placenta alters gene expression to increase nutrient supply to the fetus;26 for example, upregulation of System A transporters can increase delivery of amino acids.3, 27 There is also an increase in iron transport proteins, which absorb iron from maternal blood,28 and of placental CRH, which increases the production of maternal glucose needed to support the growing fetal brain.29 One pathway by which increased cortisol can lead to growth restriction is by interfering with CRH-driven glucose production.29 Immune function

The placenta employs a number of mechanisms that protect the embryo/fetus from infection. Genes involved in immune regulation are among the most differentially expressed30, 31 and differentially methylated32 in the placenta across different gestational ages. The human placenta is not only the source of hematopoiesis early in pregnancy, but remains a hematopoietic organ throughout gestation.33, 34 The placenta also contains a large number of Hofbauer cells (placental macrophages), which may play roles in placental angiogenesis and prevent pathogens crossing from mother to fetus.35 Exosomes and microvesicles also appear to provide protection against viruses, which may be partially attributable to transmission of members of the chromosome 19

Molecular Aspects of Placental Development 199

placenta-specific paternally expressed microRNA cluster (C19MC).36 Understanding how the placenta protects from infection is an important question in the study of preterm birth (PTB). Chorioamnionitis (CA), or intra-amniotic infection, an inflammation of the chorion and amnion usually caused by bacterial infection, is associated with the majority of extremely (T (p.Glu6Val) variant in the HBB gene, or achondroplasia caused predominantly by two pathogenic variants in FGFR3, c.1138G>A or c.1138G>C (both resulting in p.Gly380Arg substitution). For such conditions, robust assays with accurate estimates of test sensitivity and specificity as well as clinical utility can be achieved. However, single-gene disorders are more commonly caused by one of a number of different types of variants throughout a gene, or sometimes in a

CHAPTER 8

Noninvasive Prenatal Diagnosis and Screening for Monogenic Disorders Using Cell-Free DNA 323

set of related genes. For other conditions, only a subset of patients have pathogenic variants in the known disease gene(s), while for others the cause is unknown. Finally, not all have mutations detectable by sequencing of cfDNA fragments. These include structural chromosomal variants, smaller CNVs such as single exon deletions, complex insertion–deletion variants, or repeat expansions. Such issues are not unique to cfDNA testing but must be considered when these tests are introduced into the clinic.

Current status of noninvasive single-gene testing by cell-free DNA analysis Overview

The demonstration that Y chromosome-specific sequences could be amplified from maternal plasma in women who were pregnant with a male fetus provided the first evidence for the presence of fetal DNA in maternal plasma during pregnancy.10 This was soon followed by the first report in 2000 of single-gene disorder diagnosis for myotonic dystrophy,50 and several tests are now approved for clinical use.18 Table 8.1 summarizes reported experience with single-gene disorder cfDNA-based NIPD for diagnosis of autosomal dominant, autosomal recessive, and X-linked disorders in the fetus (not including sex determination or fetal RHD status). As methods become more refined newer reports are showing promising results with multiplexed assays for several diseases at once using NGS-based approaches.95 The next paragraphs highlight the reported experience with some more common conditions for which such testing has been developed and is now clinically available in some countries: fetal sex determination, Rhesus hemolytic disease of the fetus and newborn (HDFN), sickle cell disease, α- and β-thalassemia, Duchenne and Becker muscular dystrophy, CYP21A2-related congenital adrenal hyperplasia (CAH), spinal muscular atrophy (SMA), cystic fibrosis and common de novo dominant skeletal dysplasias, and other rare monogenic conditions. A wide variety of technologies have been described for delivering cfDNA-based NIPD; although the most common analysis in clinical use is now NGS, some indications are best delivered by digital PCR. Ultimately, platforms used may

vary across the globe and will also depend on an individual laboratory’s experience. However, clinical laboratories offering an accredited NIPD service must have equipment dedicated to NIPD to avoid cross-contamination. Noninvasive fetal sex determination

Cell-free DNA-based prenatal testing for fetal sex determination can be performed reliably from 7 weeks of gestation16, 18 and has value for pregnancies at increased risk of X-linked conditions, such as fragile X syndrome, hemophilia A,96 Duchenne muscular dystrophy (DMD), or X-linked adrenoleukodystrophy to help guide which pregnancies will most benefit from invasive diagnostic testing.15 Another indication is for conditions that differentially affect male and female fetuses, such as CAH.57, 97 Fetal CAH can cause virilization of the female external genitalia preventable with maternal prenatal steroid treatment.98 However, more recently, a specific noninvasive test for CAH has been developed (see later).68 Sex determination by cfDNA-based targeted testing is also helpful for the clarification of fetal ultrasound findings such as abnormalities of the fetal external genitalia or to provide additional information for diagnosing genetic conditions where genital ambiguity or sex reversal is a feature of the condition (see Chapter 12). Cell-free DNA assays for fetal sex determination usually employ real-time quantitative PCR (RT-qPCR) to identify Y chromosome-specific sequences.99 The most common determine the presence of the SRY gene or the multicopy DYS14 sequence with no significant difference in performance between these two.15 The presence of the Y chromosome sequence indicates that the fetus is male. To avoid falsely reporting a female result if Y chromosome sequences are not amplified due to technical amplification failure or low fetal fraction, the presence of fetal cfDNA must be confirmed by parallel or co-amplification of a control universal fetal cfDNA or cfRNA marker or polymorphism not present in the mother.100–102 A systematic review and meta-analysis that included 57 studies with a combined 3,524 pregnancies with male fetuses and 3,017 pregnancies with female fetuses concluded that cfDNA for fetal sex determination is reliable after 7 weeks in pregnancy.15 Nevertheless, individual tests had a failure rate of up to 5 percent

324 Genetic Disorders and the Fetus

Table 8.1 Selected studies reporting noninvasive prenatal diagnosis (NIPD) for single-gene disorders. Accredited for Condition

Method

Reference

clinical use

Autosomal dominant conditions Achondroplasia

Restriction digest

48, 130

MALDI-TOF MS

49

PCR–RED

81

QF-PCR

131

PCR–RED, NGS

90

ddPCR

86

NGS

19, 133

Multiplex PCR

134

High-resolution melting point analysis with

93

Yes – superseded by NGS Yes Yes

confirmatory melting point analysis Apert syndrome

Crouzon

AS RT PCR

211

PCR–RED

212

NGS panel

19

PCR–RED

212

Yes

NGS panel and bespoke testing

19

Huntington disease

QF-PCR

51, 213, 214

Myotonic dystrophy

Nested PCR

50

Thanatophoric dysplasia type

PCR–RED

82

Yes – superseded

PCR–RED, NGS

90

Yes

Targeted capture sequencing

72

NGS

19

Yes – panel

NGS

19

Yes – bespoke

1 (TD1) and type 2 (TD2)

Torsion dystonia

Yes

by NGS

Autosomal recessive – parents carrying different mutations Congenital adrenal

Fluorescent SNPs

57

hyperplasia Cystic fibrosis

PCR-RFLP

52

NGS for paternal exclusion

6

SnaPshot

56

MEMO fluorescent PCR

142

Digital PCR and sequencing

59

Nested PCR and restriction digestion

150

Semi-nested and nested real-time PCR for

151

Yes – superseded by RHDO

Fraser syndrome Hemoglobin E

three different mutations HB Lepore

AS-PCR

163

Leber congenital amuarosis

Denaturing HPLC

52

Polycystic kidney disease

Digital PCR and sequencing

59

Propionic acidaemia

SnaPshot; melt curve analysis.

215

Spinal muscular atrophy

NGS and RHDO

76, 139

α-Thalassemia

RT nested PCR

173

QF-PCR

176

AS RT-PCR for paternal exclusion

178

Yes

(Continued)

CHAPTER 8

Noninvasive Prenatal Diagnosis and Screening for Monogenic Disorders Using Cell-Free DNA 325

Table 8.1 (Continued) Accredited for Condition

Method

Reference

β-Thalassemia

COLD-PCR

167

AS-PCR for SNPs

164

APEX

165

Genome-wide MPS and SPRT analysis

21

AS RT fluorescent PCR for paternal exclusion

53

SABER and MS assay for paternal exclusion

160

Size selection and peptide nucleic clamp

54

Size fractionation and MALDI-TOF

161

COLD-PCR and microarray for paternal

168

clinical use

exclusion Modified fast temperature gradient

169

COLD-PCR Restriction analysis PCR

170

Quantitative genotyping and direct

171

mutation analysis Targeted NGS for paternal exclusion

89, 172

Autosomal recessive – parents carrying the same mutations Congenital adrenal

Targeted MPS and haplotype analysis

68

Targeted MPS and haplotype

69

hyperplasia analysis – hidden Markov model NGS and RHDO

19

Undergoing

Cystic fibrosis

NGS with RHDO

77, 139

Yes

Methylmalonic academia

Relative mutation dosage with ddPCR and

64

validation

parental SNP analysis Sickle cell disorder

Relative mutation dosage using digital

62

RT-PCR Pyrophosphorolysis-activated

152

polymerization (PAP) PCR-amplification with NGS

149

High-resolution melting analysis

153

Targeted MPS and RMD

156

Spinal muscular atrophy

NGS and RHDO

139

α-Thalassemia

AS RT-PCR

178

Targeted capture sequencing

180

β-Thalassemia

Semi-conductor sequencing

181

Relative mutation dosage using digital PCR

61

Targeted MPS and relative haplotype

65

Yes

dosage MPS and RMD

21

Amplicon sequencing and RMD

89

Semi-conductor sequencing

91

Targeted locus amplification and NGS

75

ddPCR

84

Relative mutation dosage using digital PCR

63

X-linked Hemophilia A and B

(Continued)

326 Genetic Disorders and the Fetus

Table 8.1 (Continued) Accredited for Condition

Method

Reference

Retinitis pigmentosa

Sequencing

58

NGS and RHDO

70, 71, 73, 139

Direct haplotyping and RHDO

140

clinical use

(X-linked) Duchenne and Becker

Yes

muscular dystrophies

This table details selected reports of NIPD for monogenic disorders. It is impossible to detail all tests done, not least since in the United Kingdom there is an accredited service for bespoke by-family testing using next-generation sequencing for a wide range of rare diseases where the mutation is known, either inherited paternally or having arisen de novo in a previous pregnancy.19 Here we also show which tests are being, to our knowledge, delivered clinically from an accredited laboratory. APEX, arrayed primer extension; AS, allele-specific; COLD, co-amplification at lower denaturation temperature; dd, droplet digital; MALDI-TOF MS, matrix-assisted laser desorption/ionization time-of-flight mass spectroscopy; MEMO, modified oligonucleotide; MPS, massively parallel sequencing; MS, mass spectroscopy; NGS, next-generation sequencing; PCR, polymerase chain reaction; QF, quantitative fluorescent; RED, restriction enzyme digest; RFLP, restriction fragment length polymorphism; RHDO, relative haplotype dosage; RMD, relative mutation dosage; RT, real time; SABER, single allele-based extension reaction; SnaPshot, single base primer extension; SNP, single-nucleotide polymorphism; SPRT, sequential probability ratio test. Source: Based on Drury S, Mason S, McKay F, et al. Implementing non-invasive prenatal diagnosis (NIPD) in a National Health Service laboratory; from dominant to recessive disorders. Adv Exp Med Biol 2016;924:71.19

in some studies, likely due to low fetal fraction,15, 16 highlighting the importance of including strategies for optimal sample collection and processing to minimize the number of inconclusive cases.103 Fetal sex determination using cfDNA analysis is available as a clinical service in several countries97, 100–102, 104 and its clinical utility has been demonstrated. An audit of UK public sector accredited laboratories found that the most common indications for testing were X-linked disorders (81.2 percent, of which 20.8 percent were for hemophilia), and CAH (11.3 percent). Of these, only 43 percent of women at risk of X-linked conditions (excluding hemophilia) and 38 percent at risk of CAH subsequently underwent invasive testing.16 A multicenter audit of fetal sex determination for 258 pregnancies at risk of CAH (134 male and 124 female fetuses) conducted in France demonstrated that prenatal steroid treatment had been avoided in 68 percent of pregnancies with a male fetus.97 The introduction of NIPD for fetal sex determination in the UK was shown to be cost-effective because of the reduction in invasive procedures and was well-received by patients because of practical and psychological benefits.105–107

Currently, determination of fetal sex is included in most of the commercial cfDNA tests for aneuploidy screening of chromosomes 13, 18, and 21 although this is not routinely reported in several countries (the Netherlands, China, United Kingdom, etc.). It is important to recognize that these approaches are not based on a targeted assay but rather on counting of cfDNA fragments, or dosage analysis of polymorphic SNPs across the X or Y chromosomes, which are affected by mosaicism. Although mosaicism can make interpretation complex, this information may have clinical utility and cannot be obtained with targeted testing for fetal sex but may also complicate analysis when fetal sex is required to target invasive testing for X-linked disorders. Fetal RHD and other blood group genotyping

The most common cause of HDFN is Rhesus D (RhD) blood group incompatibility between the mother and fetus. When a RhD-negative (RhD−) mother is pregnant with a RhD-positive (RhD+) fetus, she can develop anti-RhD antibodies which can cross the placenta and cause fetal red cell lysis, resulting in HDFN in subsequent pregnancies with a RhD+ fetus. Most cases can be prevented

CHAPTER 8

Noninvasive Prenatal Diagnosis and Screening for Monogenic Disorders Using Cell-Free DNA 327

by administration of anti-RhD immunoglobulins during pregnancy and/or after delivery to RhD− women at risk for alloimmunization along with monitoring antibody titers during pregnancy. The Rhesus blood group system is encoded by two adjacent genes on chromosome 1, the RHD gene and the RHCE gene. Most individuals with an RhD− blood type have a deletion of both copies of the RHD gene. RhD+ individuals have at least one functional copy of the RHD gene. Of these, 45 percent have two intact RHD genes and men with this genotype will always have RhD+ offspring while the other 55 percent have only one intact RHD gene, in most cases due to a heterozygous RHD gene deletion. If the father of the pregnancy of an RhD− women is heterozygous RhD+, there is a 50 percent chance the fetus will be RhD+ and at risk for HDFN. Until the development of noninvasive tests for fetal RHD genotyping, the only way to unequivocally determine if the fetus is RhD− was through amniocentesis, which is not done for routine monitoring of pregnancies at risk for sensitization, and many women would unnecessarily receive anti-RhD immunoprophylaxis to prevent alloimmunization. This motivated early development of noninvasive cfDNA-based fetal RHD genotyping.11 This can be done with a “paternal exclusion” type assay designed to detect RHD gene sequences in cfDNA since most women lack the RHD gene. Because the RHD gene structure is complex, with more than 200 rare variants and an RHD pseudogene, most assays are actually designed to analyze at least two17, 108 or more RHD exons109–111 to improve accuracy. A 2006 meta-analysis of 37 studies found an overall diagnostic accuracy of 94.8 percent for NIPD for fetal RHD genotyping, with 16 of the included studies reporting 100 percent accuracy.112 More recent large-scale studies using robust high-throughput methods suitable for routine screening reported diagnostic accuracy of greater than 99 percent from 10–11 weeks of gestation onwards,113–121 but rare false-positive, false-negative, and inconclusive results remain an issue. A false-positive result, often due to a pseudogene, would only result in unnecessary anti-RhD immunoprophylaxis, but a false-negative result could place women at risk of sensitization and potential HDFN in future pregnancies. This could be mitigated by postnatal prophylaxis if the neonate is found to be RhD+ by serology,

and since the test has >99 percent accuracy the overall risk of alloimmunization would increase minimally.117 Although the best timing for offering cfDNA-based fetal RHD genotyping by cfDNA testing is still debated,117, 122 it has now been implemented as standard practice at a national level for management of pregnancies at high risk of HDFN in a growing number of countries, largely those that did not have a robust immunoprophylaxis program already in place.108, 121, 123 It can avoid exposing women who are not at risk to unnecessary immunoprophylaxis.124 Research with women and health professionals in the United Kingdom suggests that introduction of this service would be welcomed.125 Although the test is now commercially available, it is not universally used in the United States,126 where its cost-effectiveness over routine prophylaxis has been questioned by some.127, 128 HDFN can also be due to maternal–fetal incompatibility for other Rhesus blood antigens, Rhesus C/c and Rhesus E/e, as well as incompatibility for the Kell (K/k) antigens.109 The C/c and E/e Rhesus blood types are determined by SNVs in the RHCE gene and the Kell blood type results from variants in the KEL gene, which can be tested for by sensitive quantitative PCR assays. Other technologies for predicting blood group phenotypes have also been explored. For example, massively parallel sequencing (MPS) has been used to test noninvasively for the fetal KEL genotype,129 as well as for development of a multiplexed assay for a panel of blood group genes.122

Noninvasive prenatal diagnosis of monogenic disorders Here we describe the use of cfDNA testing for the diagnosis of monogenic disorders in families at known increased risk of specific conditions, either because of a relevant family history or because of sonographic findings suggestive of a genetic condition. In this situation analysis is targeted to a known mutation or range of mutations. This testing, once validated and introduced into clinical practice, is considered diagnostic and does not require confirmation by analysis of a fetal sample obtained by invasive testing. Some of the analyses we describe are already in clinical practice

328 Genetic Disorders and the Fetus

(Table 8.1), but others, as noted, are still under development. Skeletal dysplasias

Although there are over 400 different skeletal dysplasias, the ones more commonly detected prenatally (see Chapter 20) are severe lethal conditions, such as thanatophoric dysplasia caused by autosomal dominant pathogenic variants in FGFR3, or lethal osteogenesis imperfecta, caused by pathogenic variants in COL1A1 or COL1A2. These genes are also implicated in other skeletal dysplasias that are not lethal. Achondroplasia is caused by one of two variants in FGFR3 and mutations in COL1A1 and COL1A2 can cause milder forms of osteogenesis imperfecta. Making an early diagnosis is important to differentiate these different forms from each other and from other conditions that may be recessive and have different recurrence risks or that can be associated with other health problems in addition to the skeletal dysplasia. Furthermore, many of these conditions cannot be diagnosed by early ultrasound examinations and are detected only later in gestation. These considerations have sparked strong interest in developing cfDNA-based NIPD assays for the most common skeletal dysplasias. The first studies used PCR or quantitative fluorescent PCR and restriction enzyme digestion (PCR–RED), directly or combined with a fragment size selection protocol to detect the common achondroplasia c.1138G>A mutation in FGFR3 in maternal plasma cfDNA from a patient whose fetus was known to have achondroplasia.48, 130, 131 Subsequently, size fractionation was combined with matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) with good results on two patients.49 Chitty et al. reported success with PCR–RED for cfDNA-based NIPD of fetal achondroplasia in four affected and two unaffected pregnancies that had growth restriction.81 They successfully used a similar PCR–RED approach for diagnosing thanatophoric dysplasia from maternal plasma cfDNA in three of 42 women referred for ultrasound findings consistent with severe skeletal dysplasia by detecting the c.742C>CT (p.Arg248Cys) or c.1948A>AG (p.Lys650Glu) variant in FGFR3.82 ddPCR with minisequencing of amplicons containing c.1138G nucleotide provided 100 percent sensitivity and specificity

in analysis of 26 maternal plasma samples, five of which were from women carrying a fetus with achondroplasia.86 These approaches are useful when there is predominantly one mutation that causes the condition, such as for achondroplasia. When there are multiple causative mutations in de novo conditions, such as in thanatophoric dysplasia, it becomes too complex and time consuming to test for a range of mutations using PCR-based methods that can only test for one mutation at a time. To address this, MPS was introduced to enable testing for a panel of mutations in a single assay. Comparison of PCR–RED to a newly developed NGS-based method for NIPD of achondroplasia and thanatophoric dysplasia showed in 47 maternal plasma cfDNA samples that NGS was more accurate (96.2 percent) with fewer inconclusive results.90, 132 Tests that combined multiplex PCR with NGS for 19 FGFR3 pathogenic variants that cause achondroplasia, thanatophoric dysplasias I and II and one for the five common mutation hotspots in FGFR3 yielded 100 percent sensitivity and specificity on maternal plasma samples from women with affected and unaffected pregnancies.133, 134 A prospectively conducted multicenter study that included 86 samples from women with pregnancies with achondroplasia and 65 from controls on the performance of high-resolution melting analysis (HRM) with confirmatory SNaPshot minisequencing confirmed high accuracy and suitability for introducing this test into clinical care.93 These studies remained focused on conditions caused by FGFR3 mutations, but did not address other skeletal dysplasias. Such tests are available in small panels or as bespoke analysis for individual at-risk families, for example for FGFR2 variants causing Apert syndrome or Crouzon syndrome and for COL1A1 or COL1A2 mutations causing osteogenesis imperfecta. In the United Kingdom, bespoke analysis for rarer skeletal dysplasias caused by dominant mutations that may have arisen de novo in a previous pregnancy, or which are carried by the father or for autosomal recessive variants where parents are heterozygous for different mutations is available for high-risk families.135 One group reported development of a targeted capture sequencing panel cfDNA-based NIPD that includes 16 genes associated with the most common autosomal dominant

CHAPTER 8

Noninvasive Prenatal Diagnosis and Screening for Monogenic Disorders Using Cell-Free DNA 329

and autosomal recessive lethal skeletal dysplasias: achondrogenesis IA (TRIP11), achondrogenesis IB (SLC26A2), achondrogenesis II or hypochondrogenesis (COL2A1), thanatophoric dysplasia I and II (FGFR3), short rib-polydactyly syndrome I, IIB, and III (DYNC2H1), short rib-polydactyly syndrome IIA (NEK1), fibrochondrogenesis I (COL11A1), atelosteogenesis I and III (FLNB), atelosteogenesis II (SLC26A2), perinatal osteogenesis imperfecta (COL1A1, COL1A2, CRTAP, LEPRE1, PPIB, BMP1), and hypophosphatasia (ALPL). This assay was tested on parental genomic DNA and maternal plasma cfDNA for three families with fetuses affected with a skeletal dysplasia caused by a de novo pathogenic variant, which were all detected and for two control families, which were both negative. They also were able to identify 97 percent of nonpathogenic variants that were present in the fetus but not in the mother.72 This was combined with pseudotetraploid genotyping which directly deduces the combined maternal and fetal genotype in one patient and resulted in identification of a de novo COL1A1 mutation.136 Duchenne and Becker muscular dystrophy

DMD and the milder Becker muscular dystrophy (BMD) are caused by deletions and pathogenic variants of differing severity in the X-linked DMD gene. DMD affects 1:3,500–1:6,000 male births. Boys with DMD have early normal development, followed by progressive skeletal muscle weakness of lower limbs and later also develop dilated cardiomyopathy and most become wheelchair-dependent in their early teenage years. Two-thirds of affected boys inherit the mutation from asymptomatic carrier mothers who have a 50 percent chance of having an affected son. Among the observed mutations, 60-65 percent are deletions and 5–10 percent duplications of one or more DMD exons, while 20–35 percent are SNVs or small insertions/deletions.137, 138 ECS panels currently exist that can detect the carrier mothers, but one-third of affected boys have a de novo mutation in the DMD gene, and for those carrier screening is not useful. In both scenarios, a cfDNA-based DMD NIPD test could identify affected pregnancies before the end of the first trimester.

The early noninvasive approaches were based on cfDNA-based fetal sex determination to triage women with male pregnancies for invasive testing but did not directly assay the disease-causing mutation. Beginning in 2015, several groups reported fetal DMD genotyping from cfDNA. In most of these the maternal haplotype that carries the pathogenic variant is determined first by capture NGS of the DMD gene and flanking regions containing polymorphic SNPs in parents and the affected proband, after which cfDNA-targeted sequence results are compared to the maternal haplotypes and pathogenic mutations. Prediction of the fetal genotype from maternal SNPs was 99.98 percent accurate in eight families at risk for transmitting DMD.70 Yoo et al. showed similar success in detecting deletions, duplications, and a pathogenic SNV for four mother–proband pairs.71 Parks et al. validated cfDNA analysis using an RHDO approach in seven healthy and two carrier pregnant women which was accurate in all samples with a fetal fraction >4 percent.73 NIPD using RHDO for DMD and BMD has since been introduced into clinical practice in the United Kingdom and this group have reported their experience with 35 cases at risk of these conditions.139 In five women with pregnancies at risk of BMD, four fetuses inherited the low-risk maternal haplotype and in the fifth there was a recombination event close to the familial variant, giving an inclusive result. Of the 35 pregnancies at risk of DMD, 11 were unaffected because they inherited the low-risk haplotype and 13 inherited the high-risk haplotype. Eight of these were pregnancies from known DMD carriers, but for the other five there was increased risk of parental germline mosaicism (see Chapter 14) and invasive testing was required to determine the fetal status.139 These methods still require availability of the proband DNA to determine the haplotype that carries the mutation. In 2018, Jang et al. and in 2020, Chen et al. reported success with direct targeted haplotype phasing without needing to analyze DNA from the proband or other family members in high-risk families where the mother is a carrier for DMD.79, 140 They resolved maternal haplotypes that carried the pathogenic variants by targeted linked-read sequencing of a captured region including the DMD gene and polymorphic SNPs and predicted the fetal genotype by integrating the maternal

330 Genetic Disorders and the Fetus

haplotype data with the targeted sequence data from the cfDNA to infer the fetal sex, DMD gene variants, and fetal haplotype.140 Cystic fibrosis

With an incidence of 1:2,500 livebirths in people of Northern European descent, cystic fibrosis (see Chapter 15) is one of the most common autosomal recessive disorders for which parental carrier or newborn screening is routinely offered in several countries. It is also one of the first conditions for which noninvasive testing on maternal plasma cfDNA was explored. With developments in multidisciplinary care the life expectancy of patients with severe cystic fibrosis is increasing, with a median survival now at 44 years.141 The earliest approaches focused on exclusion of the paternally inherited pathogenic variant when parents carry different mutations.6, 55, 56, 142 This approach has significant limitations since invasive testing would still be required for 50 percent of cases where the paternal mutation was detected. In addition, there is a common mutation in CFTR (p.Phe508del) that is present in approximately 4 percent of people of Northern European descent such that an estimated 47 percent of carrier couples would be ineligible for paternal mutation testing as they both carry the same mutation. However, studies in the United Kingdom indicated that NIPD would be welcomed by parents at risk of transmitting cystic fibrosis. Although 56 percent of parents said they would decline or had declined invasive testing, 94.9 percent (including those who had previously declined invasive testing) said they would choose NIPD if it were available to prepare for the birth of an affected child.6 Subsequently, Chandler et al.77 developed and validated an NGS assay of the region flanking CFTR and applied RHDO from sequencing proband and parental samples to correctly determine which alleles were inherited by the fetus for 13 cases. Following the validation and accreditation of this method, the assay was brought into clinical service in the United Kingdom with samples accepted from 9 weeks of gestation. In the first 2 years of clinical service, 36 of 38 referrals had conclusive results with known outcomes. There were two inconclusive cases, one due to a recombination event on the paternal allele and the other due to no heterozygosity around the

CFTR gene likely due to consanguinity. A similar assay has also been successfully implemented into clinical service in another UK laboratory.139 Although successful, this approach is limited by the requirement for a proband and is not suitable for use with consanguineous couples. In 2017, Vermeulen et al. reported success in implementing a proband-free RHDO approach using targeted-locus amplification of the region surrounding the CFTR gene and MPS with correct outcomes for nine pregnancies at risk of cystic fibrosis.75 Spinal muscular atrophy

SMA is an autosomal recessive group of neuromuscular disorders caused by mutations in the SMN1 gene that result in the loss of motor neurons and progressive muscle wasting.143 The severity of symptoms and age of onset varies by the type. Although rare, it is the second most common autosomal recessive condition in Northern European populations with most types apparent at birth or in the early months and years and is a significant cause of early mortality and morbidity.143 Parks and colleagues reported using RHDO in six pregnancies where parents were carriers of SMA to correctly define the fetal genotype.76 This test has since been introduced into clinical practice in the United Kingdom and this group has reported on 81 cases undergoing NIPD for SMA. They correctly identified 23 unaffected, 34 carrier, and 15 affected fetuses. Complete diagnosis was not possible in the remaining nine cases due to low fetal fraction, recombination, or lack of informativity regarding the maternal allele.139 Congenital adrenal hyperplasia

CAH is an autosomal recessive inborn error of cortisol biosynthesis. More than 90–95 percent of CAH cases are caused by steroid 21-hydroxylase deficiency caused by mutations in the CYP21A2 gene, a condition that affects 1:14,000–1:18,000 liveborns.98, 144, 145 CAH is characterized by impaired glucocorticoid and mineralocorticoid synthesis, which causes excess androgen secretion that can result in virilization of female fetuses and premature adrenarche in males.98, 144, 145 The three main clinical presentations of CAH due to 21-hydroxylase deficiency – classic salt-wasting CAH, classic simple virilizing CAH, and nonclassic

CHAPTER 8

Noninvasive Prenatal Diagnosis and Screening for Monogenic Disorders Using Cell-Free DNA 331

CAH – can be differentiated by the degree by which mutations affect protein function. Although close to 300 different CYP21A2 mutations are known, about half of all classic CAH are caused by large deletions and a common splice site mutation in intron 2 of the CYP21A2 gene.98 Prenatal treatment of CAH with maternal dexamethasone can reduce the prenatal virilization of the external genitalia of female fetuses, but to be effective such treatment has to be initiated before the onset of genital development which begins at 9 weeks of gestation.98, 144, 145 This management is controversial in view of the potential adverse effects on neuronal development and in some countries is no longer used.146 CAH and fetal gender can be diagnosed prenatally by CVS between 11 and 13 completed weeks of gestation or by amniocentesis after 15 weeks. This means that seven out of eight women with male fetuses or unaffected female fetuses were unnecessarily exposed to the side effects of dexamethasone treatment.68 Initially, cfDNA-based noninvasive screening for fetal sex was done to avoid treating male fetuses,97 but even with this, 75 percent of treated female at-risk pregnancies are actually unaffected. The initial attempts for disease-specific NIPD for CAH relied on sex determination and subsequent paternal exclusion testing to identify the fetuses that are predicted to be either unaffected or carriers for the disease.57 New and colleagues first reported their development of NIPD methods for fetal CAH in 2012,147 and in 2014 described a collaborative study wherein they successfully determined the fetal genotype of seven affected, five carriers, and two unaffected fetuses in 14 at-risk pregnancies by RHDO as early as 5–6 weeks of gestation for some.68 To achieve this, they developed and validated an MPS assay of a captured 6 Mb polymorphic region flanking CYP21A2 and deduced the parental haplotypes from sequencing the affected proband and parental samples, which allowed them to subsequently determine which alleles were inherited by the fetus.68 An additional case of successful NIPD for CAH using a haplotype-based approach on the sequenced exome was reported shortly after this.148 In 2016, Drury et al. reported on a similar assay targeting 6,700 heterozygous SNPs in a 7 Mb CYP21A2 region that was successfully validated in two sets of three families;19 validation is ongoing

to implement this test in the UK National Health Service. Ma et al. reported 14 samples from 12 independent families using an RHDO approach as early as 8 weeks of gestation.69 However, because adequate fetal fractions are required for the RHDO approach, a later gestation of at least 10–11 weeks is recommended. Hemoglobinopathies

Hemoglobinopathies, including sickle cell anemia, α-thalassemia, β-thalassemia, and anemias caused by rarer globin variants, are the most common autosomal recessive disorders worldwide (see Chapter 27), with many cases occurring in low-resource countries149 and many groups have reported development of cfDNA-based NIPD assays for these hemoglobinopathies. As with other recessive conditions, initial attempts focused on exclusion of the paternally inherited alleles,150–153 but this was soon followed by approaches to directly diagnose the fetal genotype noninvasively. However, to our knowledge this is yet to be offered routinely in an accredited molecular genetic diagnostic service. This is largely due to the fact that they are recessive conditions with multiple causative mutations, but whereby parents frequently carry the same mutation, making analysis complex.

Sickle cell anemia More than 90 percent of sickle cell anemia (see Chapter 27) is caused by biallelic inheritance of the hemoglobin β gene (HBB) gene rs334 variant (p.Glu6Val, HbS or βS) causing HbSS disease, or by co-inheritance in trans of this variant with the rs33930165 (p.Glu6Lys, HbC or βC) variant, causing milder HbSC disease. Other forms of sickle cell disease include compound heterozygous inheritance of HbS with a β-thalassemia variant or HbS with other rarer β-globin variants.95, 154 Due to the protective effect of the HbS variant against malaria, carrier frequencies for sickle cell trait (HbAS) are 1:12 for African Americans and more than 20 percent in sub-Saharan Africa with more than 200,000 births worldwide and 1,000 in the United States annually with sickle cell anemia.149, 155 In the first proof-of-principle study of cfDNA-based NIPD for sickle cell anemia, digital PCR was used for RMD for the pathogenic variant along with an assay for

332 Genetic Disorders and the Fetus

DYS14 for male pregnancies and a panel of biallelic polymorphic indel markers for female pregnancies to quantify the fetal fraction. This yielded an overall accuracy of 82 percent (37/45) in male and 75 percent (15/20) in female pregnancies and 100 percent accuracy with a fetal fraction above 7 percent.62 More recently, optimized assays have been developed.95, 149, 156 PCR amplification of HbS variant-containing amplicons combined with NGS and fetal fraction determination by RASSF1A promoter methylation analysis on 57 cfDNA samples from women between 8 and 17 weeks of gestation yielded overall sensitivity of 94 percent, specificity of 88 percent, positive predictive value (PPV) of 75 percent, and negative predictive value (NPV) of 98 percent. Specificity and PPV were 100 percent when fetal fraction was above 4 percent.149 A sensitive sequencing-based molecular counting assay with barcoded spiked-in Quantitative Counting Templates (QCTs) for cfDNA-based NIPT for various autosomal recessive single-gene disorders, including sickle cell anemia, α-thalassemia, β-thalassemia, cystic fibrosis, and SMA had >98 percent analytical sensitivity and >99 percent analytical specificity during assay development, but no actual samples from carriers of sickle cell trait were tested during validation.95 A targeted MPS-based RMD assay with incorporated unique molecular identifiers (UMIs) for NIPD of fetal sickle cell anemia, without need for paternal or proband samples, was tested on cfDNA samples from 42 pregnant HbS carriers, 15 with HbSS disease, and seven with HbSC disease at or above 8 weeks of gestational age. It yielded 100 percent clinical sensitivity and specificity with fetal fractions >4 percent.156 One sample with a fetal fraction 90–95 percent had at least one cell scorable for aneuploidy only.203 Although promising, evaluation of performance for single-gene disorder diagnosis from circulating trophoblast has been limited. One group demonstrated success with diagnosis of cystic fibrosis from circulating trophoblast cells isolated from maternal blood, but we are not aware of additional follow-up studies with this method.192 A recent study attempted diagnosis of Huntington’s disease and other single-gene disorders from circulating

CHAPTER 8

Noninvasive Prenatal Diagnosis and Screening for Monogenic Disorders Using Cell-Free DNA 337

trophoblasts with moderate success.205 Although the access to pure fetal genome is attractive, the biggest challenge with intact fetal (trophoblast) cell analysis is the small number of cells that can be recovered. In addition, significant validation is needed to determine how many cells are required to avoid the possibility of inaccurate results due

to allele dropout that has already been reported in individual cells. This will require approaches to improve cell recovery and assays that are optimized for small samples, similar to strategies developed for preimplantation genetic testing for monogenic disorders. How this will compare to cfDNA-based testing remains to be determined.

References 1. Akolekar R, Beta J, Picciarelli G, et al. Procedure-related risk of miscarriage following amniocentesis and chorionic villus sampling: a systematic review and meta-analysis. Ultrasound Obstet Gynecol 2015;45(1):16. 2. Eddleman KA, Malone FD, Sullivan L, et al. Pregnancy loss rates after midtrimester amniocentesis. Obstet Gynecol 2006;108(5):1067. 3. Salomon LJ, Sotiriadis A, Wulff CB, et al. Risk of miscarriage following amniocentesis or chorionic villus sampling: systematic review of literature and updated meta-analysis. Ultrasound Obstet Gynecol 2019;54(4):442. 4. Hill M, Compton C, Karunaratna M, et al. Client views and attitudes to non-invasive prenatal diagnosis for sickle cell disease, thalassaemia and cystic fibrosis. J Genet Couns 2014;23(6):1012. 5. Lewis C, Hill M, Chitty LS. Non-invasive prenatal diagnosis for single gene disorders: experience of patients. Clin Genet 2014;85(4):336. 6. Hill M, Twiss P, Verhoef TI, et al. Non-invasive prenatal diagnosis for cystic fibrosis: detection of paternal mutations, exploration of patient preferences and cost analysis. Prenat Diagn 2015;35(10):950. 7. ACOG Practice Bulletin No. 163. Summary: screening for fetal aneuploidy. Obstet Gynecol 2016;127(5):979. 8. Van den Veyver IB. Recent advances in prenatal genetic screening and testing. F1000Res 2016;5:2591. 9. Bianchi DW, Simpson JL, Jackson LG, et al. Fetal gender and aneuploidy detection using fetal cells in maternal blood: analysis of NIFTY I data. National Institute of Child Health and Development Fetal Cell Isolation Study. Prenat Diagn 2002;22(7):609. 10. Lo YM, Corbetta N, Chamberlain PF, et al. Presence of fetal DNA in maternal plasma and serum. Lancet 1997;350(9076):485. 11. Lo YM, Hjelm NM, Fidler C, et al. Prenatal diagnosis of fetal RhD status by molecular analysis of maternal plasma. N Engl J Med 1998;339(24):1734. 12. Fan HC, Blumenfeld YJ, Chitkara U, et al. Noninvasive diagnosis of fetal aneuploidy by shotgun sequencing

13.

14.

15.

16.

17.

18.

19.

20.

21.

22.

23.

DNA from maternal blood. Proc Natl Acad Sci U S A 2008;105(42):16266. Chiu RW, Chan KC, Gao Y, et al. Noninvasive prenatal diagnosis of fetal chromosomal aneuploidy by massively parallel genomic sequencing of DNA in maternal plasma. Proc Natl Acad Sci U S A 2008;105(51):20458. Shaw J, Scotchman E, Chandler N, et al. Non-invasive prenatal testing for aneuploidy, copy number variants and single gene disorders. Reproduction 2020;10.1530/REP-19-0591. Devaney SA, Palomaki GE, Scott JA, et al. Noninvasive fetal sex determination using cell-free fetal DNA: a systematic review and meta-analysis. JAMA 2011;306(6):627. Hill M, Finning K, Martin P, et al. Non-invasive prenatal determination of fetal sex: translating research into clinical practice. Clin Genet 2011;80(1):68. Clausen FB. Lessons learned from the implementation of non-invasive fetal RHD screening. Expert Rev Mol Diagn 2018;18(5):423. Scotchman E, Chandler NJ, Mellis R, et al. Noninvasive prenatal diagnosis of single-gene diseases: the next frontier. Clin Chem 2020;66(1):53. Drury S, Mason S, McKay F, et al. Implementing non-invasive prenatal diagnosis (NIPD) in a National Health Service laboratory; from dominant to recessive disorders. Adv Exp Med Biol 2016;924:71. Zhang J, Li J, Saucier JB, et al. Non-invasive prenatal sequencing for multiple Mendelian monogenic disorders using circulating cell-free fetal DNA. Nat Med 2019;25(3):439. Lo YM, Chan KC, Sun H, et al. Maternal plasma DNA sequencing reveals the genome-wide genetic and mutational profile of the fetus. Sci Transl Med 2010;2(61):61ra91. Chim SS, Tong YK, Chiu RW, et al. Detection of the placental epigenetic signature of the maspin gene in maternal plasma. Proc Natl Acad Sci U S A 2005;102(41):14753. Alberry M, Maddocks D, Jones M, et al. Free fetal DNA in maternal plasma in anembryonic pregnancies:

338 Genetic Disorders and the Fetus

24.

25.

26.

27.

28.

29.

30.

31.

32.

33.

34.

35.

36.

confirmation that the origin is the trophoblast. Prenat Diagn 2007;27(5):415. Mardy A, Wapner RJ. Confined placental mosaicism and its impact on confirmation of NIPT results. Am J Med Genet C Semin Med Genet 2016;172(2):118. Grati FR. Implications of feto-placental mosaicism on cell-free DNA testing: a review of a common biological phenomenon. Ultrasound Obstet Gynecol 2016;48(4):415. Malvestiti F, Agrati C, Grimi B, et al. Interpreting mosaicism in chorionic villi: results of a monocentric series of 1001 mosaics in chorionic villi with follow-up amniocentesis. Prenat Diagn 2015;35(11):1117. Lund ICB, Becher N, Christensen R, et al. Prevalence of mosaicism in uncultured chorionic villus samples after chromosomal microarray and clinical outcome in pregnancies affected by confined placental mosaicism. Prenat Diagn 2020;40(2):244. Gu S, Jernegan M, Van den Veyver IB, et al. Chromosomal microarray analysis on uncultured chorionic villus sampling can be complicated by confined placental mosaicism for aneuploidy and microdeletions. Prenat Diagn 2018;38(11):858. Palomaki GE, Kloza EM, Lambert-Messerlian GM, et al. DNA sequencing of maternal plasma to detect Down syndrome: an international clinical validation study. Genet Med 2011;13(11):913. Canick JA, Palomaki GE, Kloza EM, et al. The impact of maternal plasma DNA fetal fraction on next generation sequencing tests for common fetal aneuploidies. Prenat Diagn 2013;33(7):667. Hui L, Bianchi DW. Fetal fraction and noninvasive prenatal testing: What clinicians need to know. Prenat Diagn 2020;40(2):155. Lo YM, Zhang J, Leung TN, et al. Rapid clearance of fetal DNA from maternal plasma. Am J Hum Genet 1999;64(1):218. Hedriana H, Martin K, Saltzman D, et al. Cell-free DNA fetal fraction in twin gestations in single-nucleotide polymorphism-based noninvasive prenatal screening. Prenat Diagn 2020;40(2):179. Gromminger S, Yagmur E, Erkan S, et al. Fetal aneuploidy detection by cell-free DNA sequencing for multiple pregnancies and quality issues with vanishing twins. J Clin Med 2014;3(3):679. Hudecova I, Chiu RW. Non-invasive prenatal diagnosis of thalassemias using maternal plasma cell free DNA. Best Pract Res Clin Obstet Gynaecol 2017;39:63. Lun FM, Chiu RW, Allen Chan KC, et al. Microfluidics digital PCR reveals a higher than expected fraction of fetal DNA in maternal plasma. Clin Chem 2008;54(10):1664.

37. van Beek DM, Straver R, Weiss MM, et al. Comparing methods for fetal fraction determination and quality control of NIPT samples. Prenat Diagn 2017;37(8):769. 38. Jensen TJ, Kim SK, Zhu Z, et al. Whole genome bisulfite sequencing of cell-free DNA and its cellular contributors uncovers placenta hypomethylated domains. Genome Biol 2015;16:78. 39. Nygren AO, Dean J, Jensen TJ, et al. Quantification of fetal DNA by use of methylation-based DNA discrimination. Clin Chem 2010;56(10):1627. 40. Chan KC, Ding C, Gerovassili A, et al. Hypermethylated RASSF1A in maternal plasma: a universal fetal DNA marker that improves the reliability of noninvasive prenatal diagnosis. Clin Chem 2006;52(12):2211. 41. Lun FM, Chiu RW, Sun K, et al. Noninvasive prenatal methylomic analysis by genomewide bisulfite sequencing of maternal plasma DNA. Clin Chem 2013;59(11):1583. 42. Jiang P, Chan KC, Liao GJ, et al. FetalQuant: deducing fractional fetal DNA concentration from massively parallel sequencing of DNA in maternal plasma. Bioinformatics 2012;28(22):2883. 43. Peng XL, Jiang P. Bioinformatics approaches for fetal DNA fraction estimation in noninvasive prenatal testing. Int J Mol Sci 2017;18(2). 44. Kim SK, Hannum G, Geis J, et al. Determination of fetal DNA fraction from the plasma of pregnant women using sequence read counts. Prenat Diagn 2015;35(8):810. 45. Straver R, Oudejans CB, Sistermans EA, et al. Calculating the fetal fraction for noninvasive prenatal testing based on genome-wide nucleosome profiles. Prenat Diagn 2016;36(7):614. 46. Barrett AN, Chitty LS. Developing noninvasive diagnosis for single-gene disorders: the role of digital PCR. Methods Mol Biol 2014;1160:215. 47. Barrett AN, Xiong L, Tan TZ, et al. Measurement of fetal fraction in cell-free DNA from maternal plasma using a panel of insertion/deletion polymorphisms. PLoS One 2017;12(10):e0186771. 48. Saito H, Sekizawa A, Morimoto T, et al. Prenatal DNA diagnosis of a single-gene disorder from maternal plasma. Lancet 2000;356(9236). 49. Li Y, Page-Christiaens GC, Gille JJ, et al. Non-invasive prenatal detection of achondroplasia in size-fractionated cell-free DNA by MALDI-TOF MS assay. Prenat Diagn 2007;27(1):11. 50. Amicucci P, Gennarelli M, Novelli G, et al. Prenatal diagnosis of myotonic dystrophy using fetal DNA obtained from maternal plasma. Clin Chem 2000;46(2):301.

CHAPTER 8

Noninvasive Prenatal Diagnosis and Screening for Monogenic Disorders Using Cell-Free DNA 339

51. Gonzalez-Gonzalez Rodriguez de Alba

MC, M,

Trujillo

et

al.

MJ,

Huntington

disease-unaffected fetus diagnosed from maternal plasma using QF-PCR. Prenat Diagn 2003;23(3):232. 52. Bustamante-Aragones

A,

Vallespin

E,

Rodriguez de Alba M, et al. Early noninvasive prenatal detection of a fetal CRB1 mutation causing Leber congenital amaurosis. Mol Vis 2008;14:1388. 53. Chiu RW, Lau TK, Leung TN, et al. Prenatal exclusion of beta thalassaemia major by examination of maternal plasma. Lancet 2002;360(9338):998. 54. Li Y, Di Naro E, Vitucci A, et al. Detection of paternally inherited fetal point mutations for beta-thalassemia using size-fractionated cell-free DNA in maternal plasma. JAMA 2005;293(7):843. 55. Gonzalez-Gonzalez MC, Garcia-Hoyos M, Trujillo MJ, et al. Prenatal detection of a cystic fibrosis mutation in fetal DNA from maternal plasma. Prenat Diagn 2002;22(10):946. 56. Bustamante-Aragones

A,

Gallego-Merlo

J,

Trujillo-Tiebas MJ, et al. New strategy for the prenatal detection/exclusion of paternal cystic fibrosis mutations in maternal plasma. J Cyst Fibros 2008;7(6):505. 57. Chiu RW, Lau TK, Cheung PT, et al. Noninvasive prenatal exclusion of congenital adrenal hyperplasia by maternal plasma analysis: a feasibility study. Clin Chem 2002;48(5):778. 58. Bustamante-Aragones A, Garcia-Hoyos M, Rodriguez DEAM, et al. Detection of a paternally inherited fetal mutation in maternal plasma by the use of automated sequencing. Ann N Y Acad Sci. 2006;1075:108. 59. Lench N, Barrett A, Fielding S, et al. The clinical implementation of non-invasive prenatal diagnosis for single-gene disorders: challenges and progress made. Prenat Diagn 2013;33(6):555. 60. Chiu EKL, In Hui WW, Chiu RWK. cfDNA screening and diagnosis of monogenic disorders – where are we heading? Prenat Diagn 2018;38(1):52. 61. Lun FM, Tsui NB, Chan KC, et al. Noninvasive prenatal diagnosis of monogenic diseases by digital size selection and relative mutation dosage on DNA in maternal plasma. Proc Natl Acad Sci U S A 2008;105(50):19920. 62. Barrett AN, McDonnell TC, Chan KC, et al. Digital PCR analysis of maternal plasma for noninvasive detection of sickle cell anemia. Clin Chem 2012;58(6):1026. 63. Tsui NB, Kadir RA, Chan KC, et al. Noninvasive prenatal diagnosis of hemophilia by microfluidics digital PCR analysis of maternal plasma DNA. Blood 2011;117(13):3684.

64. Gu W, Koh W, Blumenfeld YJ, et al. Noninvasive prenatal diagnosis in a fetus at risk for methylmalonic acidemia. Genet Med 2014;16(7):564. 65. Lam KW, Jiang P, Liao GJ, et al. Noninvasive prenatal diagnosis of monogenic diseases by targeted massively parallel sequencing of maternal plasma: application to beta-thalassemia. Clin Chem 2012;58(10):1467. 66. Fan HC, Gu W, Wang J, et al. Non-invasive prenatal measurement of the fetal genome. Nature 2012;487(7407):320. 67. Kitzman JO, Snyder MW, Ventura M, et al. Noninvasive whole-genome sequencing of a human fetus. Sci Transl Med 2012;4(137):137ra76. 68. New MI, Tong YK, Yuen T, et al. Noninvasive prenatal diagnosis of congenital adrenal hyperplasia using cell-free fetal DNA in maternal plasma. J Clin Endocrinol Metab 2014;99(6):E1022. 69. Ma D, Yuan Y, Luo C, et al. Noninvasive prenatal diagnosis of 21-hydroxylase deficiency using target capture sequencing of maternal plasma DNA. Sci Rep 2017;7(1):7427. 70. Xu Y, Li X, Ge HJ, et al. Haplotype-based approach for noninvasive prenatal tests of Duchenne muscular dystrophy using cell-free fetal DNA in maternal plasma. Genet Med 2015;17(11):889. 71. Yoo SK, Lim BC, Byeun J, et al. Noninvasive prenatal diagnosis of duchenne muscular dystrophy: comprehensive genetic diagnosis in carrier, proband, and fetus. Clin Chem 2015;61(6):829. 72. Dan S, Yuan Y, Wang Y, et al. Non-invasive prenatal diagnosis of lethal skeletal dysplasia by targeted capture sequencing of maternal plasma. PLoS One 2016;11(7):e0159355. 73. Parks M, Court S, Cleary S, et al. Non-invasive prenatal diagnosis of Duchenne and Becker muscular dystrophies by relative haplotype dosage. Prenat Diagn 2016;36(4):312. 74. Zeevi DA, Altarescu G, Weinberg-Shukron A, et al. Proof-of-principle rapid noninvasive prenatal diagnosis of autosomal recessive founder mutations. J Clin Invest 2015;125(10):3757. 75. Vermeulen C, Geeven G, de Wit E, et al. Sensitive monogenic noninvasive prenatal diagnosis by targeted haplotyping. Am J Hum Genet 2017;101(3):326. 76. Parks M, Court S, Bowns B, et al. Non-invasive prenatal diagnosis of spinal muscular atrophy by relative haplotype dosage. Eur J Hum Genet 2017;25(4):416. 77. Chandler NJ, Ahlfors H, Drury S, et al. Noninvasive prenatal diagnosis for cystic fibrosis: implementation, uptake, outcome, and implications. Clin Chem 2020;66(1):207.

340 Genetic Disorders and the Fetus

78. Che H, Villela D, Dimitriadou E, et al. Noninvasive prenatal diagnosis by genome-wide haplotyping of cell-free plasma DNA. Genet Med 2020;22(5):962. 79. Chen M, Chen C, Huang X, et al. Noninvasive prenatal diagnosis for Duchenne muscular dystrophy based on the direct haplotype phasing. Prenat Diagn 2020;40(8):918. 80. Hui WW, Jiang P, Tong YK, et al. Universal haplotype-based noninvasive prenatal testing for single gene diseases. Clin Chem 2017;63(2):513. 81. Chitty LS, Griffin DR, Meaney C, et al. New aids for the non-invasive prenatal diagnosis of achondroplasia: dysmorphic features, charts of fetal size and molecular confirmation using cell-free fetal DNA in maternal plasma. Ultrasound Obstet Gynecol 2011;37(3):283. 82. Chitty LS, Khalil A, Barrett AN, et al. Safe, accurate, prenatal diagnosis of thanatophoric dysplasia using ultrasound and free fetal DNA. Prenat Diagn 2013;33(5):416. 83. Caswell RC, Snowsill T, Houghton JAL, et al. Noninvasive fetal genotyping by droplet digital PCR to identify maternally inherited monogenic diabetes variants. Clin Chem 2020;66(7):958. 84. Camunas-Soler J, Lee H, Hudgins L, et al. Noninvasive prenatal diagnosis of single-gene disorders by use of droplet digital PCR. Clin Chem 2018;64(2):336. 85. Perlado S, Bustamante-Aragones A, Donas M, et al. Fetal genotyping in maternal blood by digital PCR: towards NIPD of monogenic disorders independently of parental origin. PLoS One 2016;11(4):e0153258. 86. Orhant L, Anselem O, Fradin M, et al. Droplet digital PCR combined with minisequencing, a new approach to analyze fetal DNA from maternal blood: application to the non-invasive prenatal diagnosis of achondroplasia. Prenat Diagn 2016;36(5):397. 87. Chang MY, Kim AR, Kim MY, et al. Development of novel noninvasive prenatal testing protocol for whole autosomal recessive disease using picodroplet digital PCR. Sci Rep 2016;6:37153. 88. De Franco E, Caswell R, Houghton JA, et al. Analysis of cell-free fetal DNA for non-invasive prenatal diagnosis in a family with neonatal diabetes. Diabet Med 2017;34(4):582. 89. Xiong L, Barrett AN, Hua R, et al. Non-invasive prenatal diagnostic testing for beta-thalassaemia using cell-free fetal DNA and next generation sequencing. Prenat Diagn 2015;35(3):258. 90. Chitty LS, Mason S, Barrett AN, et al. Non-invasive prenatal diagnosis of achondroplasia and thanatophoric dysplasia: next-generation sequencing allows for a safer, more accurate, and comprehensive approach. Prenat Diagn 2015;35(7):656.

91. Saba L, Masala M, Capponi V, et al. Non-invasive prenatal diagnosis of beta-thalassemia by semiconductor sequencing: a feasibility study in the sardinian population. Eur J Hum Genet 2017;25(5):600. 92. Xiong L, Barrett AN, Hua R, et al. Non-invasive prenatal testing for fetal inheritance of maternal beta-thalassaemia mutations using targeted sequencing and relative mutation dosage: a feasibility study. BJOG 2018;125(4):461. 93. Vivanti AJ, Costa JM, Rosefort A, et al. Optimal non-invasive diagnosis of fetal achondroplasia combining ultrasonography and circulating cell-free fetal DNA analysis. Ultrasound Obstet Gynecol 2019;53(1):87. 94. Dello Russo C, Cesta A, Longo S, et al. Validation of extensive next-generation sequencing method for monogenic disorder analysis on cell-free fetal DNA: noninvasive prenatal diagnosis. J Mol Diagn 2019;21(4):572. 95. Tsao DS, Silas S, Landry BP, et al. A novel high-throughput molecular counting method with single base-pair resolution enables accurate single-gene NIPT. Sci Rep 2019;9(1):14382. 96. Hill M, Compton C, Lewis C, et al. Determination of foetal sex in pregnancies at risk of haemophilia: a qualitative study exploring the clinical practices and attitudes of health professionals in the United Kingdom. Haemophilia 2012;18(4):575. 97. Tardy-Guidollet V, Menassa R, Costa JM, et al. New management strategy of pregnancies at risk of congenital adrenal hyperplasia using fetal sex determination in maternal serum: French cohort of 258 cases (2002–2011). J Clin Endocrinol Metab 2014;99(4):1180. 98. Speiser PW, Arlt W, Auchus RJ, et al. Congenital adrenal hyperplasia due to steroid 21-hydroxylase deficiency: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 2018;103(11):4043. 99. Zimmermann B, El-Sheikhah A, Nicolaides K, et al. Optimized real-time quantitative PCR measurement of male fetal DNA in maternal plasma. Clin Chem 2005;51(9):1598. 100. Hill M, Lewis C, Jenkins L, et al. Implementing noninvasive prenatal fetal sex determination using cell-free fetal DNA in the United Kingdom. Expert Opin Biol Ther 2012;12(Suppl 1):S119. 101. Scheffer PG, van der Schoot CE, Page-Christiaens GC, et al. Reliability of fetal sex determination using maternal plasma. Obstet Gynecol 2010;115(1):117. 102. Bustamante-Aragones A, Rodriguez de Alba M, Gonzalez-Gonzalez C, et al. Foetal sex determination in maternal blood from the seventh week of gestation

CHAPTER 8

103.

104.

105.

106.

107.

108.

109.

110.

111.

112.

113.

114.

Noninvasive Prenatal Diagnosis and Screening for Monogenic Disorders Using Cell-Free DNA 341

and its role in diagnosing haemophilia in the foetuses of female carriers. Haemophilia 2008;14(3):593. Barrett AN, Zimmermann BG, Wang D, et al. Implementing prenatal diagnosis based on cell-free fetal DNA: accurate identification of factors affecting fetal DNA yield. PLoS One 2011;6(10):e25202. Hyett JA, Gardener G, Stojilkovic-Mikic T, et al. Reduction in diagnostic and therapeutic interventions by non-invasive determination of fetal sex in early pregnancy. Prenat Diagn 2005;25(12):1111. Hill M, Taffinder S, Chitty LS, et al. Incremental cost of non-invasive prenatal diagnosis versus invasive prenatal diagnosis of fetal sex in England. Prenat Diagn 2011;31(3):267. Lewis C, Hill M, Skirton H, et al. Non-invasive prenatal diagnosis for fetal sex determination: benefits and disadvantages from the service users’ perspective. Eur J Hum Genet 2012;20(11):1127. Lewis C, Hill M, Skirton H, et al. Fetal sex determination using cell-free fetal DNA: service users’ experiences of and preferences for service delivery. Prenat Diagn 2012;32(8):735. Daniels G, Finning K, Martin P, et al. Noninvasive prenatal diagnosis of fetal blood group phenotypes: current practice and future prospects. Prenat Diagn 2009;29(2):101. Finning K, Martin P, Summers J, et al. Fetal genotyping for the K (Kell) and Rh C, c, and E blood groups on cell-free fetal DNA in maternal plasma. Transfusion 2007;47(11):2126. Grande M, Ordonez E, Cirigliano V, et al. Clinical application of midtrimester non-invasive fetal RHD genotyping and identification of RHD variants in a mixed-ethnic population. Prenat Diagn 2013;33(2):173. Tynan JA, Angkachatchai V, Ehrich M, et al. Multiplexed analysis of circulating cell-free fetal nucleic acids for noninvasive prenatal diagnostic RHD testing. Am J Obstet Gynecol 2011;204(3):251 e1. Geifman-Holtzman O, Grotegut CA, Gaughan JP. Diagnostic accuracy of noninvasive fetal Rh genotyping from maternal blood – a meta-analysis. Am J Obstet Gynecol 2006;195(4):1163. Clausen FB, Christiansen M, Steffensen R, et al. Report of the first nationally implemented clinical routine screening for fetal RHD in D− pregnant women to ascertain the requirement for antenatal RhD prophylaxis. Transfusion 2012;52(4):752. Rouillac-Le Sciellour C, Puillandre P, Gillot R, et al. Large-scale pre-diagnosis study of fetal RHD genotyping by PCR on plasma DNA from RhD-negative pregnant women. Mol Diagn 2004;8(1):23.

115. Van der Schoot CE, Soussan AA, Koelewijn J, et al. Non-invasive antenatal RHD typing. Transfus Clin Biol 2006;13(1-2):53. 116. Finning K, Martin P, Summers J, et al. Effect of high throughput RHD typing of fetal DNA in maternal plasma on use of anti-RhD immunoglobulin in RhD negative pregnant women: prospective feasibility study. BMJ 2008;336(7648):816. 117. Chitty LS, Finning K, Wade A, et al. Diagnostic accuracy of routine antenatal determination of fetal RHD status across gestation: population based cohort study. BMJ 2014;349:g5243. 118. de Haas M, Thurik FF, van der Ploeg CP, et al. Sensitivity of fetal RHD screening for safe guidance of targeted anti-D immunoglobulin prophylaxis: prospective cohort study of a nationwide programme in the Netherlands. BMJ 2016;355:i5789. 119. Muller SP, Bartels I, Stein W, et al. The determination of the fetal D status from maternal plasma for decision making on Rh prophylaxis is feasible. Transfusion 2008;48(11):2292. 120. Wikman AT, Tiblad E, Karlsson A, et al. Noninvasive single-exon fetal RHD determination in a routine screening program in early pregnancy. Obstet Gynecol 2012;120(2 Pt 1):227. 121. Johnson JA, MacDonald K, Clarke G, et al. No. 343-routine non-invasive prenatal prediction of fetal RHD genotype in Canada: the time is here. J Obstet Gynaecol Can 2017;39(5):366. 122. Clausen FB. Integration of noninvasive prenatal prediction of fetal blood group into clinical prenatal care. Prenat Diagn 2014;34(5):409. 123. Soothill P, Finning K, Latham T, et al. Use of cffDNA to avoid administration of anti-D to pregnant women when the fetus is RhD-negative: implementation in the NHS. BJOG 2015;122(12):1682. 124. Kent J, Farrell AM, Soothill P. Routine administration of Anti-D: the ethical case for offering pregnant women fetal RHD genotyping and a review of policy and practice. BMC Pregnancy Childbirth 2014;14:87. 125. Oxenford K, Silcock C, Hill M, et al. Routine testing of fetal Rhesus D status in Rhesus D negative women using cell-free fetal DNA: an investigation into the preferences and information needs of women. Prenat Diagn 2013;33(7):688. 126. ACOG Practice Bulletin No. 192: Management of alloimmunization during pregnancy. Obstet Gynecol 2018;131(3):e82. 127. Moise KJ. Selected use of antenatal Rhesus-immune globulin based on free fetal DNA. BJOG 2015;122(12):1687.

342 Genetic Disorders and the Fetus

128. Moise KJ, Jr., Hashmi SS, Markham K, et al. Cell free fetal DNA to triage antenatal rhesus immune globulin: is it really cost-effective in the United States? Prenat Diagn 2019;39(3):238. 129. Rieneck K, Bak M, Jonson L, et al. Next-generation sequencing: proof of concept for antenatal prediction of the fetal Kell blood group phenotype from cell-free fetal DNA in maternal plasma. Transfusion 2013;53(11 Suppl 2):2892. 130. Li Y, Holzgreve W, Page-Christiaens GC, et al. Improved prenatal detection of a fetal point mutation for achondroplasia by the use of size-fractionated circulatory DNA in maternal plasma – case report. Prenat Diagn 2004;24(11):896. 131. Lim JH, Kim MJ, Kim SY, et al. Non-invasive prenatal detection of achondroplasia using circulating fetal DNA in maternal plasma. J Assist Reprod Genet 2011;28(2):167. 132. Drury S, Hill M, Chitty LS. Cell-free fetal DNA testing for prenatal diagnosis. Adv Clin Chem 2016;76:1. 133. Ren Y, Zhao J, Li R, et al. Noninvasive prenatal test for FGFR3-related skeletal dysplasia based on next-generation sequencing and plasma cell-free DNA: test performance analysis and feasibility exploration. Prenat Diagn 2018;38(11):821. 134. Terasawa S, Kato A, Nishizawa H, et al. Multiplex PCR in noninvasive prenatal diagnosis for FGFR3-related disorders. Congenit Anom (Kyoto) 2019;59(1):4. 135. Hayward J, Chitty LS. Beyond screening for chromosomal abnormalities: Advances in non-invasive diagnosis of single gene disorders and fetal exome sequencing. Semin Fetal Neonatal Med 2018;23(2):94. 136. Yin X, Du Y, Zhang H, et al. Identification of a de novo fetal variant in osteogenesis imperfecta by targeted sequencing-based noninvasive prenatal testing. J Hum Genet 2018;63(11):1129. 137. Aartsma-Rus A, Van Deutekom JC, Fokkema IF, et al. Entries in the Leiden Duchenne muscular dystrophy mutation database: an overview of mutation types and paradoxical cases that confirm the reading-frame rule. Muscle Nerve 2006;34(2):135. 138. Prior TW, Bridgeman SJ. Experience and strategy for the molecular testing of Duchenne muscular dystrophy. J Mol Diagn 2005;7(3):317. 139. Young E, Bowns B, Gerrish A, et al. Clinical service delivery of noninvasive prenatal diagnosis by relative haplotype dosage for single-gene disorders. J Mol Diagn 2020;22(9):1151. 140. Jang SS, Lim BC, Yoo S-K, et al. Targeted linked-read sequencing for direct haplotype phasing of maternal DMD alleles: a practical and reliable method for noninvasive prenatal diagnosis. Sci Rep 2018;8(1):8678.

141. Cystic Fibrosis Trust. UK Cystic Fibrosis Registry Annual Data Report 2018. August 2019. 142. Guissart C, Dubucs C, Raynal C, et al. Non-invasive prenatal diagnosis (NIPD) of cystic fibrosis: an optimized protocol using MEMO fluorescent PCR to detect the p.Phe508del mutation. J Cyst Fibros 2017;16(2):198. 143. Prior TW, Leach ME, Finanger E. Spinal muscular atrophy. 2000 Feb 24 (updated 2019 Nov 14). In: GeneReviews [Internet]. Seattle, WA: University of Washington, Seattle; 1993–2020. https://www.ncbi.nlm.nih .gov/books/NBK1352/ 144. White PC. Update on diagnosis and management of congenital adrenal hyperplasia due to 21-hydroxylase deficiency. Curr Opin Endocrinol Diabetes Obes 2018;25(3):178. 145. Simpson JL, Rechitsky S. Prenatal genetic testing and treatment for congenital adrenal hyperplasia. Fertil Steril 2019;111(1):21. 146. Chitty LS, Chatelain P, Wolffenbuttel KP, et al. Prenatal management of disorders of sex development. J Pediatr Urol 2012;8(6):576. 147. New MI, Abraham M, Yuen T, et al. An update on prenatal diagnosis and treatment of congenital adrenal hyperplasia. Semin Reprod Med 2012;30(5):396. 148. Ma D, Ge H, Li X, et al. Haplotype-based approach for noninvasive prenatal diagnosis of congenital adrenal hyperplasia by maternal plasma DNA sequencing. Gene 2014;544(2):252. 149. Cutts A, Vavoulis DV, Petrou M, et al. A method for noninvasive prenatal diagnosis of monogenic autosomal recessive disorders. Blood 2019;134(14):1190. 150. Fucharoen G, Tungwiwat W, Ratanasiri T, et al. Prenatal detection of fetal hemoglobin E gene from maternal plasma. Prenat Diagn 2003;23(5):393. 151. Tungwiwat W, Fucharoen G, Fucharoen S, et al. Application of maternal plasma DNA analysis for noninvasive prenatal diagnosis of Hb E-beta-thalassemia. Transl Res 2007;150(5):319. 152. Phylipsen M, Yamsri S, Treffers EE, et al. Non-invasive prenatal diagnosis of beta-thalassemia and sickle-cell disease using pyrophosphorolysis-activated polymerization and melting curve analysis. Prenat Diagn 2012;32(6):578. 153. Yenilmez ED, Tuli A, Evruke IC. Noninvasive prenatal diagnosis experience in the Cukurova Region of Southern Turkey: detecting paternal mutations of sickle cell anemia and beta-thalassemia in cell-free fetal DNA using high-resolution melting analysis. Prenat Diagn 2013;33(11):1054. 154. Ware RE, de Montalembert M, Tshilolo L, et al. Sickle cell disease. The Lancet 2017;390(10091):311.

CHAPTER 8

Noninvasive Prenatal Diagnosis and Screening for Monogenic Disorders Using Cell-Free DNA 343

155. Ojodu J, Hulihan MM, Pope SN, et al. Incidence of sickle cell trait--United States, 2010. MMWR Morb Mortal Wkly Rep 2014;63(49):1155. 156. van Campen J, Silcock L, Yau S, et al. A novel non-invasive prenatal sickle cell disease test for all at-risk pregnancies. Br J Haematol 2020;190(1):119. 157. Chakravorty S, Rees D. Commentary on sickle cell non-invasive prenatal testing article. Br J Haematol 2020;190(1):20. 158. Galanello R, Cao A. Gene test review. Alphathalassemia. Genet Med 2011;13(2):83. 159. Cao A, Galanello R. Beta-thalassemia. Genet Med 2010;12(2):61. 160. Ding C, Chiu RW, Lau TK, et al. MS analysis of single-nucleotide differences in circulating nucleic acids: application to noninvasive prenatal diagnosis. Proc Natl Acad Sci U S A 2004;101(29):10762. 161. Li Y, Di Naro E, Vitucci A, et al. Size fractionation of cell-free DNA in maternal plasma improves the detection of a paternally inherited beta-thalassemia point mutation by MALDI-TOF mass spectrometry. Fetal Diagn Ther 2009;25(2):246. 162. Galbiati S, Foglieni B, Travi M, et al. Peptide-nucleic acid-mediated enriched polymerase chain reaction as a key point for non-invasive prenatal diagnosis of beta-thalassemia. Haematologica 2008;93(4):610. 163. Lazaros L, Hatzi E, Bouba I, et al. Non-invasive prenatal detection of paternal origin hb lepore in a male fetus at the 7th week of gestation. Fetal Diagn Ther 2006;21(6):506. 164. Papasavva T, Kalakoutis G, Kalikas I, et al. Noninvasive prenatal diagnostic assay for the detection of beta-thalassemia. Ann N Y Acad Sci 2006;1075:148. 165. Papasavva T, Kalikas I, Kyrri A, et al. Arrayed primer extension for the noninvasive prenatal diagnosis of beta-thalassemia based on detection of single nucleotide polymorphisms. Ann N Y Acad Sci 2008;1137:302. 166. Chan K, Yam I, Leung KY, et al. Detection of paternal alleles in maternal plasma for non-invasive prenatal diagnosis of beta-thalassemia: a feasibility study in southern Chinese. Eur J Obstet Gynecol Reprod Biol 2010;150(1):28. 167. Galbiati S, Brisci A, Lalatta F, et al. Full COLD-PCR protocol for noninvasive prenatal diagnosis of genetic diseases. Clin Chem 2011;57(1):136. 168. Galbiati S, Monguzzi A, Damin F, et al. COLD-PCR and microarray: two independent highly sensitive approaches allowing the identification of fetal paternally inherited mutations in maternal plasma. J Med Genet 2016;53(7):481.

169. Byrou S, Makrigiorgos GM, Christofides A, et al. Fast temperature-gradient COLD PCR for the enrichment of the paternally inherited SNPs in cell free fetal DNA; an application to non-invasive prenatal diagnosis of beta-thalassaemia. PLoS One 2018;13(7):e0200348. 170. Liu S, Chen L, Zhang X, et al. Primer-introduced restriction analysis polymerase chain reaction method for non-invasive prenatal testing of beta-thalassemia. Hemoglobin 2015;39(1):18. 171. Breveglieri G, Travan A, D’Aversa E, et al. Postnatal and non-invasive prenatal detection of beta-thalassemia mutations based on Taqman genotyping assays. PLoS One 2017;12(2):e0172756. 172. Papasavva T, van Ijcken WF, Kockx CE, et al. Next generation sequencing of SNPs for non-invasive prenatal diagnosis: challenges and feasibility as illustrated by an application to beta-thalassaemia. Eur J Hum Genet 2013;21(12):1403. 173. Tungwiwat W, Fucharoen S, Fucharoen G, et al. Development and application of a real-time quantitative PCR for prenatal detection of fetal alpha(0)-thalassemia from maternal plasma. Ann N Y Acad Sci 2006;1075:103. 174. Sirichotiyakul S, Charoenkwan P, Sanguansermsri T. Prenatal diagnosis of homozygous alpha-thalassemia-1 by cell-free fetal DNA in maternal plasma. Prenat Diagn 2012;32(1):45. 175. Pornprasert S, Sukunthamala K, Kunyanone N, et al. Analysis of real-time PCR cycle threshold of alpha-thalassemia-1 Southeast Asian type deletion using fetal cell-free DNA in maternal plasma for noninvasive prenatal diagnosis of Bart’s hydrops fetalis. J Med Assoc Thai. 2010;93(11):1243. 176. Ho SS, Chong SS, Koay ES, et al. Noninvasive prenatal exclusion of haemoglobin Bart’s using foetal DNA from maternal plasma. Prenat Diagn 2010;30(1):65. 177. Li X, Yang T, Li CS, et al. Prenatal detection of thalassemia by cell-free fetal DNA (cffDNA) in maternal plasma using surface enhanced Raman spectroscopy combined with PCR. Biomed Opt Express 2018;9(7):3167. 178. Yan TZ, Mo QH, Cai R, et al. Reliable detection of paternal SNPs within deletion breakpoints for non-invasive prenatal exclusion of homozygous alpha-thalassemia in maternal plasma. PLoS One 2011;6(9):e24779. 179. Ge H, Huang X, Li X, et al. Noninvasive prenatal detection for pathogenic CNVs: the application in alpha-thalassemia. PLoS One 2013;8(6):e67464. 180. Wang W, Yuan Y, Zheng H, et al. A pilot study of noninvasive prenatal diagnosis of alpha- and beta-thalassemia with target capture sequencing of

344 Genetic Disorders and the Fetus

cell-free fetal DNA in maternal blood. Genet Test Mol Biomarkers 2017;21(7):433. 181. Yang J, Peng CF, Qi Y, et al. Noninvasive prenatal

193. Beaudet AL. Using fetal cells for prenatal diagnosis: History and recent progress. Am J Med Genet C Semin Med Genet 2016;172(2):123.

detection of hemoglobin Bart hydrops fetalis via mater-

194. Breman AM, Chow JC, U’Ren L, et al. Evidence for

nal plasma dispensed with parental haplotyping using

feasibility of fetal trophoblastic cell-based noninvasive

the semiconductor sequencing platform. Am J Obstet Gynecol 2020;222(2):185 e1.

prenatal testing. Prenat Diagn 2016;36(11):1009. 195. Normand E, Qdaisat S, Bi W, et al. Comparison of

182. ACOG. Practice advisory: cell-free DNA to screen

three whole genome amplification methods for detec-

for single-gene disorders, 2019, updated Feb 21,

tion of genomic aberrations in single cells. Prenat Diagn

2019. https://www.acog.org/Clinical-Guidance-andPublications/Practice-Advisories/Cell-free-DNA-toScreen-for-Single-Gene-Disorders 183. Lewis C, Hill M, Chitty L. Non-invasive prenatal diagnosis for single gene disorders: experience of patients. Clin Genet 2014;85(4):336. 184. Hill M, Oteng-Ntim E, Forya F, et al. Preferences for prenatal diagnosis of sickle-cell disorder: a discrete choice experiment comparing potential service users and health-care providers. Health Expect 2017;20(6):1289. 185. National Health Service. UK national genomic test directory, 2018, updated 20 March 2019. https://www .england.nhs.uk/publication/national-genomic-testdirectories/ 186. Wilkie AOM, Goriely A. Gonadal mosaicism and non-invasive prenatal diagnosis for ‘reassurance’ in sporadic paternal age effect (PAE) disorders. Prenat Diagn 2017;37(9):946. 187. Deans Z, Hill M, Chitty LS, et al. Non-invasive prenatal testing for single gene disorders: exploring the ethics. Eur J Hum Genet 2013;21(7):713. 188. Skirton H, Goldsmith L, Chitty LS. An easy test but a hard decision: ethical issues concerning non-invasive prenatal testing for autosomal recessive disorders. Eur J Hum Genet 2015;23(8):1004.

2016;36(9):823. 196. Kolvraa S, Singh R, Normand EA, et al. Genome-wide copy number analysis on DNA from fetal cells isolated from the blood of pregnant women. Prenat Diagn 2016;36(12):1127. 197. Vestergaard EM, Singh R, Schelde P, et al. On the road to replacing invasive testing with cell-based NIPT: five clinical cases with aneuploidies, microduplication, unbalanced structural rearrangement or mosaicism. Prenat Diagn 2017;37(11):1120. 198. Huang CE, Ma GC, Jou HJ, et al. Noninvasive prenatal diagnosis of fetal aneuploidy by circulating fetal nucleated red blood cells and extravillous trophoblasts using silicon-based nanostructured microfluidics. Mol Cytogenet 2017;10:44. 199. Chen F, Liu P, Gu Y, et al. Isolation and whole genome sequencing of fetal cells from maternal blood towards the ultimate non-invasive prenatal testing. Prenat Diagn 2017;37(13):1311. 200. Wei X, Ao Z, Cheng L, et al. Highly sensitive and rapid isolation of fetal nucleated red blood cells with microbead-based selective sedimentation for non-invasive prenatal diagnostics. Nanotechnology 2018;29(43):434001. 201. Vossaert L, Wang Q, Salman R, et al. Reliable detection

189. Westgren M, Gotherstrom C. Stem cell transplantation

of subchromosomal deletions and duplications using

before birth – a realistic option for treatment of osteo-

cell-based noninvasive prenatal testing. Prenat Diagn

genesis imperfecta? Prenat Diagn 2015;35(9):827.

2018;38(13):1069.

190. Verhoef TI, Hill M, Drury S, et al. Non-invasive pre-

202. Huang ZW, Fong CY, Gauthaman K, et al. Biol-

natal diagnosis (NIPD) for single gene disorders: cost

ogy of human primitive erythroblasts for applica-

analysis of NIPD and invasive testing pathways. Prenat

tion in noninvasive prenatal diagnosis. Prenat Diagn

Diagn 2016;36(7):636.

2018;38(9):673.

191. Jenkins LA, Deans ZC, Lewis C, et al. Delivering an

203. Vossaert L, Wang Q, Salman R, et al. Validation stud-

accredited non-invasive prenatal diagnosis service for

ies for single circulating trophoblast genetic testing as

monogenic disorders and recommendations for best

a form of noninvasive prenatal diagnosis. Am J Hum

practice. Prenat Diagn 2018;38(1):44.

Genet 2019;105(6):1262.

192. Mouawia H, Saker A, Jais JP, et al. Circulating tro-

204. Huang Y, Situ B, Huang L, et al. Nondestructive identi-

phoblastic cells provide genetic diagnosis in 63 fetuses

fication of rare trophoblastic cells by endoplasmic retic-

at risk for cystic fibrosis or spinal muscular atrophy.

ulum staining for noninvasive prenatal testing of mono-

Reprod Biomed Online 2012;25(5):508.

genic diseases. Adv Sci (Weinh) 2020;7(7):1903354.

CHAPTER 8

Noninvasive Prenatal Diagnosis and Screening for Monogenic Disorders Using Cell-Free DNA 345

205. Cayrefourcq L, Vincent MC, Pierredon S, et al. Single circulating fetal trophoblastic cells eligible for non invasive prenatal diagnosis: the exception rather than the rule. Sci Rep 2020;10(1):9861. 206. Imudia AN, Suzuki Y, Kilburn BA, et al. Retrieval of trophoblast cells from the cervical canal for prediction of abnormal pregnancy: a pilot study. Hum Reprod 2009;24(9):2086. 207. Bolnick AD, Fritz R, Jain C, et al. Trophoblast retrieval and isolation from the cervix for noninvasive, first trimester, fetal gender determination in a carrier of congenital adrenal hyperplasia. Reprod Sci 2016;23(6):717. 208. Jain CV, Kadam L, van Dijk M, et al. Fetal genome profiling at 5 weeks of gestation after noninvasive isolation of trophoblast cells from the endocervical canal. Sci Transl Med 2016;8(363):363re4. 209. Kadam L, Jain C, Kohan-Ghadr HR, et al. Endocervical trophoblast for interrogating the fetal genome and assessing pregnancy health at five weeks. Eur J Med Genet 2019:103690. 210. Pfeifer I, Benachi A, Saker A, et al. Cervical trophoblasts for non-invasive single-cell genotyping and prenatal diagnosis. Placenta 2016;37:56.

211. Au PK, Kwok YK, Leung KY, et al. Detection of the S252W mutation in fibroblast growth factor receptor 2 (FGFR2) in fetal DNA from maternal plasma in a pregnancy affected by Apert syndrome. Prenat Diagn 2011;31(2):218. 212. Raymond FL, Whittaker J, Jenkins L, et al. Molecular prenatal diagnosis: the impact of modern technologies. Prenat Diagn 2010;30(7):674. 213. Bustamante-Aragones A, Trujillo-Tiebas MJ, Gallego-Merlo J, et al. Prenatal diagnosis of Huntington disease in maternal plasma: direct and indirect study. Eur J Neurol 2008;15(12):1338. 214. Gonzalez-Gonzalez MC, Garcia-Hoyos M, Trujillo-Tiebas MJ, et al. Improvement in strategies for the non-invasive prenatal diagnosis of Huntington disease. J Assist Reprod Genet 2008;25(9–10):477. 215. Bustamante-Aragones A, Perez-Cerda C, Perez B, et al. Prenatal diagnosis in maternal plasma of a fetal mutation causing propionic acidemia. Mol Genet Metab 2008;95(1–2):101.

9

Amniocentesis, Chorionic Villus Sampling, and Fetal Blood Sampling Anthony O. Odibo University of South Florida, Tampa, FL, USA

Introduction

Amniocentesis

Amniocentesis, chorionic villus sampling (CVS), and, to a lesser extent, fetal blood sampling are the most common prenatal diagnostic techniques. Amniocentesis was first used in Germany in the early 1880s to treat polyhydramnios.1, 2 Earlier uses of amniocentesis included aiding in evaluating the fetus to localizing the placenta, and even as a method of pregnancy termination by injecting hypertonic saline into the amniotic cavity.3, 4 In 1950, Alvarez performed amniocentesis to assess fetal wellbeing.5 The use of amniocentesis increased rapidly in the 1950s, when spectrophotometric analysis of bilirubin proved valuable in monitoring fetuses with Rh isoimmunization.5, 6 In the mid-1950s amniocentesis for exclusively genetic indications evolved when several investigators demonstrated that fetal sex could be determined by X-chromatin analysis of amniotic fluid cells (AFCs).7–9 Over the next few years, several reports of successful diagnosis of a wide variety of chromosomal and metabolic disorders were published, helping to establish amniocentesis as an integral part of modern obstetric care.10–16 This chapter addresses current techniques and the safety of genetic amniocentesis, CVS, and fetal blood sampling. Indications and methods of prenatal diagnosis are considered in detail throughout this text.17–22

Prerequisites

Ideally, couples should have the opportunity to discuss their genetic risks and available antenatal testing options before pregnancy.17, 21, 23 This may necessitate genetic counseling and the counselor should elicit an accurate history, confirm the diagnosis of any abnormality in question, be aware of diagnostic capabilities, and be cognizant of psychologic defenses (e.g. denial, guilt reactions, and blame) engendered during genetic counseling. Couples must understand the risks of amniocentesis itself, the accuracy and limitations of antenatal diagnosis, the time required before results become available, technical problems potentially necessitating a second amniocentesis, and the rare possibility of an inability to make a diagnosis. Amniocentesis should be performed only by an obstetrician who is experienced in this procedure, has high-quality ultrasonography available, and has access to a laboratory with experience in performing prenatal diagnostic studies.20, 24, 25 Only obstetricians should perform the procedure because the operator must always be prepared to deal with the potential complications of the procedure. According to the American College of Obstetricians and Gynecologists (ACOG), if an abnormality is detected and the couple elects to terminate the pregnancy, the obstetrician must

Genetic Disorders and the Fetus: Diagnosis, Prevention, and Treatment, Eighth Edition. Edited by Aubrey Milunsky and Jeff M. Milunsky. © 2021 Aubrey Milunsky and Jeff M. Milunsky. Published 2021 by John Wiley & Sons Ltd.

346

CHAPTER 9

Amniocentesis, Chorionic Villus Sampling, and Fetal Blood Sampling 347

either perform the abortion or refer the family to a provider who will act on their request.26 Timing

Amniocentesis is typically performed between 15th and 16th weeks of gestation, when the ratio of viable to nonviable cells is greatest compared with procedures performed later in gestation.27 In addition, the uterus is accessible by an abdominal approach and contains sufficient amniotic fluid (AF) (200–250 mL) to permit 20–30 mL to be aspirated safely. Since most couples wait for the findings of detailed anatomic ultrasound survey prior to amniocentesis, the majority of procedures are now performed between the 18th and 20th weeks of gestation. Early amniocentesis performed before 14 weeks of gestation is discussed below. Transvaginal amniocentesis is only of historical interest, because of its technical difficulty and because of associated infection and spontaneous abortion.28 Technique

Amniocentesis for genetic diagnosis is typically performed in an outpatient facility. A careful ultrasonographic examination is performed and a needle insertion site is selected. The needle is inserted, employing concurrent ultrasound guidance to correspond to the location of the optimal pocket of AF while avoiding the fetus. Although Tabor et al.29 reported that transplacental needle insertion increased the risk of the procedure, this has not been confirmed by others.30, 31 The umbilical cord and its insertion site should be especially identified and avoided. The maternal bowel and bladder should also be located and avoided. A local anesthetic (e.g. 2–3 mL of 1 percent xylocaine) may or may not be used; however, local anesthesia, including the use of creams or subfreezing the needle, does not appear to affect the level of pain of the procedure.32–35 It has been suggested that needle insertion through the upper third of the uterus is less painful than insertion through the lower two-thirds; however, data to substantiate this claim are limited.36 Counseling before amniocentesis should emphasize that the actual pain and anxiety experienced during the procedure are significantly lower than expected.37, 38 After the maternal skin has been cleansed with an iodine-based and/or alcohol-based solution, sterile

drapes are placed around the needle insertion site to help maintain an aseptic field. A disposable 22-gauge spinal needle with stylet is most frequently used and recommended. During the entire procedure, two-dimensional, real-time ultra-sonographic monitoring with continuous visualization of the needle should be performed. Use of four-dimensional ultrasound guidance has been suggested, but there are no objective data to indicate improved outcomes.39 Ultrasound gel is applied adjacent to the insertion site, and a real-time transducer is held in position by an assistant such that the ultrasound beam is directed at a 15–20∘ angle from the parallel of the planned needle track (Figure 9.1). After assurance that the needle is in its proper location, the stylet is moved and a 10 or 20 mL syringe attached. The tip is typically more easily identified on removal of the stylet. If freely flowing AF is not obtained on aspiration, the needle must be repositioned with stylet in place. The first several milliliters are theoretically most likely to contain maternal cells from blood vessels, the abdominal wall, or the myometrium; therefore, this initial sample is usually discarded or set aside for AF 𝛼-fetoprotein (AFAFP) assay. Twenty to thirty milliliters of AF are aspirated into sterile, disposable plastic syringes, although as little as 3–5 mL of AF has been shown to suffice for reliable prenatal cytogenetic results.40,41 Maternal cell contamination appears to occur more frequently in genetic amniocentesis samples that are obtained by physicians who perform 40 years (5.1 percent). Women with a history of vaginal bleeding during the current pregnancy also had a higher fetal loss rate (6.5 percent) compared with controls (2.8 percent). Women with a history of previous spontaneous abortions/terminations had a fetal loss rate of 8 percent, compared with a 2.8 percent loss rate among controls.

First- and Second-Trimester Evaluation of Risk (FaSTER) Trial Research Consortium In 2006, Eddleman et al. reported the procedurerelated fetal loss rate after midtrimester amniocentesis using the database from the NICHD-sponsored multicenter First- and Second-Trimester Evaluation of Risk (FaSTER) trial designed to compare first-trimester Down syndrome screening with nuchal translucency, pregnancy-associated plasma protein A, and free β-human chorionic gonadotropin (β-hCG) to second-trimester screening with AFP, hCG, unconjugated estriol, and inhibin A.148 Among a total of 35,003 patients who were enrolled in the FaSTER trial, 3,096 underwent midtrimester amniocentesis (study group) and 31,907 did not (control group). The rate of fetal loss 1,300

Huntington disease

AD

CAG

6–36



35–121

Kennedy disease (spinal bulbar muscular atrophy)

X

CAG

12–34



40–62

Machado–Joseph disease

AD

CAG

13–36



68–79

Myotonic dystrophy type 1

AD

CTG

5–37



50 to >2,000

Myotonic dystrophy type 2d

AD

CCTG

500

Spinocerebellar ataxia type 1

AD

CAG

6–39



41–81

Spinocerebellar ataxia type 2

AD

CAG

15–29



35–59

Spinocerebellar ataxia type 6

AD

CAG

4–16



21–27

Spinocerebellar ataxia type 7

AD

CAG

4–18



37–130

Spinocerebellar ataxia type 8

AD

CTG

16–37



>90 >19,000

Spinocerebellar ataxia type 10e

AD

ATTCT

10–22



Spinocerebellar ataxia type 12

AD

CAG

7–28



66–78

Spinocerebellar ataxia type 17

AD

CAG

27–44



>45 >2,500

Spinocerebellar ataxia type 31e

AD

TGGAA

Not detected



Spinocerebellar ataxia type 36f

AD

GGCCTG

3–14



650 to 2,500

Spinocerebellar ataxia type 37e

AD

ATTTC

Not detected



31–75

a AD,

autosomal dominant; AR, autosomal recessive; X, X-linked.

b Variable

ranges reported and overlapping sizes may occur.

c Mutation

may not involve an expansion.

d Expansion

involves four nucleotides.

e Expansion

involves five nucleotides.

f Expansion

involves six nucleotides.

Sources: La Spada AR, Taylor JP. Repeat expansion disease: progress and puzzles in disease pathogenesis. Nat Rev Genet 2010;1:247;119 López Castel A, Cleary JD, Pearson CE. Repeat instability as the basis for human diseases and as a potential target for therapy. Nat Rev Mol Cell Biol 2010;11:165;120 Mootha VV, Gong X, Ku HC, et al. Association and familial segregation of CTG18.1 trinucleotide repeat expansion of TCF4 gene in Fuchs’ endothelial corneal dystrophy. Invest Ophthalmol Vis Sci 2014;55:33;121 Rodriguez CM, Todd PK. New pathologic mechanisms in nucleotide repeat expansion disorders. Neurobiol Dis 2019;130:104515;122 Tian Y, Wang JL, Huang W, et al. Expansion of human-specific GGC repeat in neuronal intranuclear inclusion disease-related disorders. Am J Hum Genet 2019;105:166;123 Swinnen B, Robberecht W, Van Den Bosch L. RNA toxicity in non-coding repeat expansion disorders. EMBO J 2020;39:e101112;124 Ueyama M, Nagai Y. Repeat expansion disease models. Adv Exp Med Biol 2018;1076:63;125 Babi´c Leko M, Župunski V, Kirincich J, et al. Molecular mechanisms of neurodegeneration related to C9orf72 hexanucleotide repeat expansion. Behav Neurol 2019;2019:2909168;126 Goodman LD, Bonini NM. Repeat-associated non-AUG (RAN) translation mechanisms are running into focus for GGGGCC-repeat associated ALS/FTD. Prog Neurobiol 2019;183:101697;127 LaCroix AJ, Stabley D, Sahraoui R, et al. GGC repeat expansion and exon 1 methylation of XYLT1 Is a common pathogenic variant in Baratela-Scott syndrome. Am J Hum Genet 2019;104:35;128 Stoyas CA, La Spada AR. The CAG-polyglutamine repeat diseases: a clinical, molecular, genetic, and pathophysiologic nosology. Handb Clin Neurol 2018;147:143;129 Saade JS, Xing C, Gong X, et al. Instability of TCF4 triplet repeat expansion with parent-child transmission in Fuchs’ endothelial corneal dystrophy. Invest Ophthalmol Vis Sci 2018;59:4065;130 and Nguyen L, Cleary JD, Ranum LPW. Repeat-associated non-ATG translation: molecular mechanisms and contribution to neurological disease. Annu Rev Neurosci 2019;42:227.131

582 Genetic Disorders and the Fetus

anticipation has also been reported.134 Clinical presentations include psychosis, hallucinations, delusions, bulbar involvement, dementia, and motor disability. More widespread knowledge of the manifestations of C9orf72 expansion mutations and the 50 percent risk of transmission is likely to invoke a greater use of prenatal diagnosis. Although technical problems in assays for the C9orf72 hexanucleotide expansions have been noted,136 Rollinson et al.137 have reported a repeat expansion mutation initially undetectable by Southern blotting. In the two brothers studied they identified a 10-base pair deletion adjacent to the expansion that interfered with the genotyping. In spinocerebellar ataxia (SCA) type 10, an autosomal dominant neurodegenerative disorder, interruptions in the pentanucleotide (ATTCT) repeat expansions are associated with a paradoxical contraction in intergenerational repeat size.138 An inverse correlation has been noted between the expansion size and the age of onset.138 For many of the SCAs (Table 14.2), clinical onset is in the reproductive years, posing personal challenges for affected individuals (self-extinction) considering prenatal diagnosis. It is important to recall that unstable repeat expansions occur in the following SCA types: 1, 2, 3, 6, 7, 8, 10, 12, 17, 31, and 36, while conventional mutations characterize other SCAs, including types 5, 11, 13, 14, 15, 20, 23, 27, 28, 35, and 38.139, 140 SCA type 37 is caused by an intronic insertion of the pentanucleotide ATTTC in between two polymorphic ATTTT tracks in the DAB1 gene.141 Long-range PCR with Sanger sequencing should reliably detect the pathogenic ATTTC repeat insertion that is typically 31–75 repeats.142 However, repeat primed PCR may also detect the ATTTT repeats and should not be used due to the potential for false-negative/positive results. Autosomal dominant myotonic muscular dystrophy (DM1) (discussed in Chapter 1), due to a CTG expansion, occurs in 1 in 8,000, while the more common DM2 (1 in 1,830 in Finland) is often undiagnosed. DM2, due to a CTTG expansion, is milder, rarely has anticipation, and does not cause congenital myotonic dystrophy.143 Distinction between these two types is important because prenatal diagnosis is unlikely to be requested in DM2. Children with congenital myotonic muscular

dystrophy (DM1) are almost exclusively born to affected mothers. Fuchs endothelial corneal dystrophy is a common genetically heterogeneous disorder with the late-onset type 3 affecting approximately 4 percent of the population older than 40 years, resulting in impaired vision. This type is caused by a heterozygous CTG expansion in the TCF4 gene.144 An expanded allele >40 repeats increases the risk of the disorder (>30-fold increase).121 More severe disease is correlated with higher expansions145 similar to other triplet repeat disorders. However, given the documented incomplete penetrance146 it is not well suited for prenatal diagnosis. There are at least nine heritable disorders due to the expansion of polyalanine tracts.147 For those that are autosomal recessive or X-linked in origin, prenatal diagnosis is feasible. Only rarely would there be similar options for those transmitted as autosomal dominant. Similar to the unstable repeat expansions, clinical severity increases with the length of the expanded polyalanine tract in some of these genes (e.g. PHOX2B).148 Mosaicism

Somatic mosaicism is the consequence of postzygotic de novo mutations occurring in a portion of all cells that constitute our organs and is almost certainly underestimated in its frequency. Moreover, due to a broad range of factors, because of subtle phenotypic changes, technical limitations, and tissue differences, mosaicism is often undetected or undiagnosed. Indeed, many mutations that may be present in all cells may affect some tissues more than others. De novo mutations likely arise in the sperm or ovum of a parent in whom the fateful change is not determinable. Once transmitted, the mutations are found in all tissues of the offspring. Disease-causing mutations may occur during mitotic cell divisions, leading ultimately to offspring who are mosaic. Somatic mutations occur frequently in the preimplantation stage or during early embryogenesis.149, 150 It is surprising that somatic mosaicism is not more commonly encountered, as 48–90 percent of embryos studied were mosaic for at least chromosomal abnormalities.151 Further to the discussion in Chapter 1, the distinction between somatic and gonadal mosaicism may often depend upon the sensitivity of the assay used. Standard clinical assays missed detection of

CHAPTER 14

mutant alleles subsequently identified by a specialized assay with greater sensitivity.152–156 Campbell et al.157 studied 100 families with children who had genomic disorders due to rare deletion CNVs thought to be de novo. In that study, four cases of low-level somatic mosaicism were found in a parent’s blood, clearly (but unwittingly) increasing future risk. Of course, limited mosaicism in another tissue may not be known (or discoverable), yet be in association with concomitant undeterminable gonadal mosaicism. De novo somatic mutations are now recognized as a cause of neuronal migration and brain overgrowth disorders as well as intellectual disability, autism spectrum disorders, and epileptic encephalopathies.158 The occurrence and description of transmissible somatic mosaicism is well known for a wide range of genetic disorders (see Chapter 1). The extant issues of limited assay reliability in the face of low percentage mosaicism in blood introduces a significant reservation to conclusions that infer gonadal mosaicism for many described genetic disorders (Table 14.3). Clinical caution is therefore advised when considering a genetic disorder as sporadic or due to gonadal mosaicism. The risks of recurrence are likely to be significantly higher if undetected somatic mosaicism is present. Most new mutations are paternal in origin and increase in frequency with advancing age.271 Moreover, recurrence risk, according to in silico analysis, depends on the parent of origin, with paternal age being an important variable that modifies risk.272, 273 Young fathers who transmit a mutant gamete appear to have higher recurrence risks. In addition, it is has been demonstrated that de novo deletions and insertions that are associated with autism spectrum disorder are predominantly paternally derived.274 Somatic mutations are more common for deletions (more often related to a severe phenotype) than for other types of mutations in neurofibromatosis type 1.275 Microdeletions in the NF1 gene are reportedly associated with somatic mosaicism in 40 percent of cases.276 In neurofibromatosis type 2, estimates indicate a high frequency of somatic mosaicism (25–30 percent).277, 278 A study of 10,362 consecutive patients by exon-targeted high-resolution whole-genome oligonucleotide array concluded that somatic chromosomal mosaicism occurred in 0.55 percent of cases.279

Molecular Genetics and Prenatal Diagnosis 583

In disorders with significant frequencies of somatic mosaicism, pursuit of the diagnosis may require examination of other tissue besides blood. A good example is the Cornelia de Lange syndrome, which is characterized by distinctive craniofacial dysmorphism, growth restriction, intellectual disability, and limb malformations.280 This autosomal dominant or X-linked congenital malformation syndrome is known to arise as a consequence of mutations in seven different genes.281 Limited sensitivity of Sanger sequencing to detect somatic mosaicism occurring at a 10–20 percent level prompted the recommendation to examine skin fibroblasts or buccal epithelial cells, enabling confirmation of mosaicism.282–284 A similar example has occurred in Costello syndrome (facial dysmorphism and cardiovascular, skin, and musculoskeletal abnormalities along with tumor predisposition) in which no detectable mutations in blood DNA were observed. Subsequently the typical HRAS mutation was found in repeated buccal epithelial cells.285 More routine utilization of NGS technologies has yielded increased detection of somatic mosaicism in many disorders.155, 156 As each base pair is interrogated from dozens to hundreds of times, detection of lower levels of mosaicism (even as low as 0.4 percent) is now possible.286 Some monogenic disorders exist only in the mosaic state (e.g. Proteus syndrome), suggesting that a full complement of mutations would be lethal. Prenatal diagnosis of Proteus syndrome by ultrasound has been made following findings that included a cystic abdominal mass and malpositioned fingers287 and abdominal and pelvic cystic lymphangioma.288 Of these disorders (such as the Maffuci syndrome – overgrowth of vascular, lymphatic, soft and bone tissues) most probably exist primarily in the mosaic state with germline mutations being lethal.275 Somatic PIK3CA pathogenic variants have been described in a variety of overgrowth disorders of the brain and body segments.289–293 The phenomenon of revertant mosaicism is not likely to complicate efforts of parental or fetal studies aimed at prenatal diagnosis. Fetal skin biopsy to diagnose epidermolysis bullosa has now been effectively replaced by gene sequencing, thereby circumventing the possibility of sampling normal skin patches, which occur in about a third of these patients.294

584 Genetic Disorders and the Fetus

Table 14.3 Selected monogenic disorders with reported gonadal mosaicism Disorder

Inheritance

Reference

Achondrogenesis type II

AD

159

Achondroplasia

AD

160

Acro-cardio-facial syndrome

AR

161

Adrenoleukodystrophy

X-L rec

162, 163

Albright hereditary osteodystrophy

AD

164

𝛼-Thalassemia mental retardation syndrome

X-L

165

Alport syndrome

X-L, AR, AD

166, 167

Amyloid polyneuropathy

AD

168

Aniridia

AD

169

Ankyloblepharon–ectodermal defects–cleft

AD

170

lip/palate (AEC syndrome) Apert syndrome

AD

171

Arboleda–Tham Syndrome

AD

172

Bainbridge–Ropers syndrome

AD

173

Becker muscular dystrophy

X-L rec

174

Cantu syndrome

AD

175

Cardiofaciocutaneous syndrome

AD

176

Cerebellar ataxia with progressive macular

AD

177

dystrophy (SCA7) CHARGE syndrome

AD

178

Chondrodysplasia punctata

X-L rec

179

Coffin–Lowry syndrome

X-L dom

180

Congenital contractural arachnodactyly

AD

181

Congenital fibrosis of extraocular muscles

AD

182

Congenital central hypoventilation

AD

183, 184

syndrome Conradi–Hunnermann–Happle syndrome

X-L dom

185

Cornelia de Lange syndrome

AD

186

Costello syndrome

AD

187

Crouzon syndrome

AD

188

Danon disease

X-L

189

Dejerine–Sotas syndrome (HNSN III) with

AD

190

stomatocytosis Duchenne muscular dystrophy

X-L rec

191, 192

Dyskeratosis congenita

X-L

193

EEC syndrome (ectrodactyly, ectodermal

AD

194, 195

dysplasia, orofacial clefts) Epidermolysis bullosa, dystrophic

AR

196

Fabry disease

X-L rec

197

Facioscapulohumeral muscular dystrophy

AD

198

Factor X deficiency

AR

199

Familial adenomatous polyposis

AD

200, 201

Familial focal segmental glomerulosclerosis

AD

202

Familial hypertrophic cardiomyopathy

AD

203

Fibrodysplasia ossificans progressiva

AD

204

Fragile X syndrome (deletion type)

X-L

205

Fraser syndrome

AD

206

Glass syndrome

AD

207

Gonadal dysgenesis

AD

208 (Continued)

CHAPTER 14

Molecular Genetics and Prenatal Diagnosis 585

Table 14.3 (Continued) Disorder

Inheritance

Reference

Hartsfield syndrome

AD

209

Hemophilia B

X-L rec

210, 211

Hereditary angioedema (C1 inhibitor

AD, AR

212

Hereditary hemorrhagic telangiectasia

AD

213

Herlitz junctional epidermolysis bullosa

AR

214

Holt–Oram syndrome

AD

215

Hunter syndrome

X-L rec

216

Hypophosphatemic rickets

X-L dom

217

Incontinentia pigmenti

X-L dom

218

deficiency)

Karsch–Neugebauer syndrome

AD

219

Keratitis–ichthyosis deafness syndrome

AD

220

L1 syndrome

X-L rec

221

Lissencephaly (males); “subcortical band

X-L rec

222

heterotopia” (almost all females) LMNA-associated muscular dystrophy

AD

223

Malan syndrome

AD

224

Megalencephaly syndrome

AD

225

Microdeletion 1p36

AD

226

Microdeletion 15q11–q13 (Angelman

AD

227

Microdeletion 19p13.13

AD

228

Myotubular myopathy

X-L rec

229, 230

Nemaline myopathy

AD, AR

231

Neurodegeneration with brain iron

X-L

232

AD

233

AD

234

syndrome)

accumulation Neurodevelopmental disorder with involuntary movements (NEDIM) Neurodevelopmental disorder with severe motor impairment and absent language (NEDMIAL) Neurofibromatosis type 1

AD

235, 236, 237

Neurofibromatosis type 2

AD

238

Noonan syndrome

AD

239, 240

Oculocerebrorenal syndrome of Lowe

X-L

241

Oculofaciocardiodental

X-L dom

242

Ornithine transcarbamylase deficiency

X-L rec

243

Osteogenesis imperfecta

AD

244

Osteopathia striata

X-L dom

245

Otopalatodigital syndrome

X-L dom

246

Pallister–Hall syndrome

AD

247

Polycystic kidney disease

AD

248

Polycythemia–paraganglioma syndrome

AD

249

Pseudoachondroplasia

AD

250

Renal coloboma syndrome

AD

251

Retinoblastoma

AD

252, 253

Rett syndrome

X-L dom, AD

254, 255, 256

Rhabdoid tumor predisposition

AD

257, 258

Schizophrenia susceptibility

AD

259

syndrome/congenital cataracts

(Continued)

586 Genetic Disorders and the Fetus

Table 14.3 (Continued) Disorder

Inheritance

Reference

Severe combined immunodeficiency disease

X-L rec

260

Shprintzen–Goldberg syndrome

AD

261

Smith–Kingsmore syndrome

AD

262

Spondyloepimetaphyseal dysplasia

AD

263

Sotos syndrome

AD

264

Tuberous sclerosis

AD

265, 266

von Willebrand disease (type 2b)

X-L rec

267

Waardenburg syndrome

AD

268, 269

Wiskott–Aldrich syndrome

X-L rec

270

AD, autosomal dominant; AR, autosomal recessive; X-L rec, X-linked recessive; X-L dom, X-linked dominant.

Imprinting and uniparental disomy

Epigenetic modifications that enable parent-oforigin preferential gene expression result in genomic imprinting.295 DNA methylation at CpG dinucleotides is the predominant modification that enables detection by methylation-sensitive PCR approaches to characterize this group of imprinting disorders. Imprinted domains containing more than 100 genes are clustered on several chromosomes, including 6, 7, 11, 14, 15, and 20.296–299 In the early embryo the previous methylation at these locations is reset to establish new parent-specific imprints.300, 301 Deletions and duplications of these imprinted regions can result in imprinting disorders. Uniparental disomy (see Chapter 1) in which two copies of an imprinted region are derived from one parent also results in these disorders (Table 14.4). Indications for the prenatal diagnosis of these disorders may include nonhomologous Robertsonian translocations involving chromosome 14 and/or 15 (risk significantly less than 1 percent),329, 330 the detection of chromosomal mosaicism,331 or the use of assisted reproductive technologies (ART).332–336 Genotype–phenotype correlations

Well-defined genotype–phenotype correlations exist for multiple disorders (see Chapter 1). For example, specific FBN1 mutations result in isolated ectopia lentis in patients that do not meet Ghent criteria for Marfan syndrome.337 More severe disease and earlier age of onset of neonatal Marfan syndrome has been documented for mutations in exons 24–32 of FBN1.338 Different types of

mutations within the same gene may lead to the same disorder with striking differences in severity. For the COL3A1 gene, significant difference in morbidity and mortality in vascular Ehlers–Danlos syndrome is associated with missense mutations, in contrast to truncating/frameshift/deletion mutations.339 Furthermore, compound heterozygosity for different types of mutations (i.e. leading to severe or mild effects on the protein) typify the resultant CBAVD (CFTR gene).340–342 For monogenic disorders many studies have identified modifier genes including microRNAs that influence the expression and severity of disease.343–345 Complicating matters further is the well-known variable expressivity and penetrance issues seen even within families affected by common genetic disorders.346 Complex genetic mechanisms exist, including digenic inheritance,347 which often confound accurate genetic counseling in these cases. Whole-exome studies have revealed multiple causative alleles for many conditions.348 Some of these conditions have differential tissue-specific results of specific classes of mutations. In addition, there are deleterious mutations that individually are necessary but not sufficient to cause disease without additional mutations.349 Hence, except for specific well-characterized known mutations, it is very difficult to predict a precise phenotype from genotype in the prenatal setting. Additional cautions and considerations

Extreme caution attends the use of all prenatal diagnostic modalities, given the potential for irreversible decisions concerning continuation (or

CHAPTER 14

Molecular Genetics and Prenatal Diagnosis 587

Table 14.4 Examples of imprinting and human disease Syndrome

Chromosomal location

Parental origin

Selected references

Albright hereditary osteodystrophy

20q13.32

Maternal

302

Angelman syndrome

15q11-q13

Maternal

303, 304

Autism

15q11-q13

Maternal

305

Beckwith–Wiedemann syndrome

11p15.5

Paternal

306, 307

Birk–Barel syndrome

8q24

Maternal

308

Congenital hyperinsulinism

11p15

Maternal

309

Congenital myotonic muscular dystrophy

19q13.3

Maternal

310

Early embryonic failure

21

Maternal

311

Familial paraganglioma

11q23

Paternal

312

Hereditary myoclonus–dystonia

7q21

Paternal

313

Intellectual disability and dysmorphism

14

Paternal

314

Intrauterine and postnatal growth restriction

7

Maternal

315

Intrauterine growth restriction or miscarriage

16

Maternal

316

Intrauterine growth restriction

11p15.5

Paternal

317

MatUPD14-like (Temple syndrome)

14

Maternal

318

PatUPD14-like syndrome

14

Paternal

319

Prader–Willi syndrome

15q11-q13

Paternal

320 321

Progressive osseous heteroplasia

20q13.3

Paternal

Pseudohypoparathyroidism 1a

20q13.3

Maternal

302

Pseudohypoparathyroidism 1b

20q13.3

Paternal

322

Pseudohypoparathyroidism

20q13.32

Paternal

323

Russell–Silver syndrome

7p11.2

Maternal

324

11p15

Maternal

325

11p15

Paternal

317

Schaaf–Yang syndrome

15q11.2-q12

Paternal

326

Short stature

14

Maternal

327

Transient neonatal diabetes

6q24.2

Paternal

328

not) of pregnancy. Complex considerations are necessary, with examples of a broad range (but commonly shared) of key caveats and cautions emphasized in the following discussion. There is a wide range of genetic disorders that result in cardiomyopathy. Causal genes and their mutations have been identified for many of these disorders, with more genes to be discovered. In families even with adult onset of hypertrophic cardiomyopathy, prenatal diagnosis could be a consideration, given that at least 16 genes have been identified, each encoding different components of the sarcomere. However, thus far, mutations have been recognized in only up to 60 percent of those affected and who have a family history.350 Complicating matters further, about 6 percent of affected individuals may possess more than one gene variant or, worse still, variants in more than one gene.351 Any consideration of prenatal

diagnosis for dilated cardiomyopathy requires precise molecular diagnosis focusing on the more than two dozen genes and their causal mutations. Again a wide range of primarily myopathic disorders can be considered, including the typical myopathies, limb-girdle muscular dystrophy type 2J, Barth syndrome, and the dystrophinopathies. Prenatal diagnosis can proceed only with precise determination of the culprit mutation. In addition to Noonan syndrome, discussed earlier, another RASopathy is the cardiofaciocutaneous syndrome (see Table 14.1). In this condition, hypertrophic cardiomyopathy, cellular dysplasias, septal defects, and dysrhythmias also occur. Thus far, four genes are known to be causally related to this syndrome with BRAF gene mutations accounting for about 75 percent of cases and the MAP2K2 for about 25 percent of cases. Given that neurological and variable intellectual disability is

588 Genetic Disorders and the Fetus

a constant in those with this disorder, prenatal diagnosis becomes a consideration. As the vast majority of cases occur from a de novo mutation, consideration of prenatal diagnosis would arise only if midtrimester ultrasound signs raised the question of a RASopathy. Craniofacial features may be similar to those described in Noonan syndrome, which, if suspected by ultrasound study, should lead to the offer of RASopathy molecular analysis (discussed earlier). An arrhythmogenic right ventricular cardiomyopathy may present in early childhood or at the mean age of diagnosis at 31 years. At least 14 genes are known to be casually related to this serious/lethal disorder.352 Where autosomal dominant inheritance applies (the frequency of de novo mutations is not known) and given the serious nature of this disorder, prenatal diagnosis or PGT would be a consideration once a precise mutation has been identified. Complicating this potential issue further is the fact that digenic inheritance might occur in a different but related arrhythmogenic cardiomyopathy gene, or the offspring could inherit one or two mutations.353 Digenic inheritance (heterozygous mutations in two separate autosomal recessive genes that only together result in the disorder) is well documented in families with hearing loss (e.g. connexin-26 and connexin-30)354, 355 and retinitis pigmentosa.356 These not-so-rare circumstances need careful attention when providing genetic counseling and prenatal diagnosis. Furthermore, triallelic inheritance has been described in Bardet–Biedl syndrome.357 The prenatal diagnosis and PGT of neurologic disorders were discussed in Chapters 1 and 2, respectively. Experience in the Netherlands has indicated that most individuals at 50 percent risk of inheriting Huntington disease (the paradigm for presymptomatic or predictive testing) prefer not knowing their possible presymptomatic status.358 It is likely that patients in the rest of the Western world have generally similar views. The Dutch investigators showed that, in their experience, exclusion prenatal diagnosis or exclusion PGT were acceptable for all the couples they studied. They emphasized yet again the importance of comprehensive and timely nondirective counseling, as well as professional and psychological support during and after the entire process of testing.

The study of cohorts of patients with neurodevelopmental and psychiatric disorders, using a wide range of technologies, has yielded inherited and de novo CNVs and rearrangements as well as single-nucleotide variants (SNVs).79, 359–364 Specifically, whole-exome/genome sequencing has revealed a plethora of de novo pathogenic variants, especially in autism.365–369 The study of many of these disorders by microarray previously revealed what was thought to be singularly pathogenic CNVs responsible for complex phenotypes such as autism. More recent studies have supported a multiple-hit model of autism spectrum disorders in some cases requiring both a pathogenic SNV as well as a susceptibility CNV to yield the specific clinical phenotype.370–372 Hence, care is needed in the prenatal diagnosis and counseling of these families that may only have one potential pathogenic variant in a multiple-hit model. In some of these cases, a maternally inherited CNV when combined with a paternally derived de novo single-nucleotide pathogenic variant may result in a specific neuropsychiatric phenotype, whereas inheritance of the maternal CNV alone may not. Thrombocytopenia-absent radius (TAR) syndrome is a disorder in which neonatal thrombocytopenia is present in combination with various radial ray defects (typically bilateral absence of the radii) with preservation of the thumbs. Various additional anomalies may be present in this autosomal recessive disorder. Most patients have a chromosome 1q21.1 deletion373 on one allele encompassing the RBM8A gene in trans with a typically hypomorphic (partial loss of gene activity) heterozygous RBM8A allele.374 About 50–75 percent of affected individuals have inherited the deletion from an unaffected carrier parent. The carrier frequency of one of the hypomorphic alleles is about 3 percent.375 Interestingly, TAR syndrome does not occur when two hypomorphic alleles in trans are detected. The rare occurrence of a homozygous chromosome 1q21.1 deletion is thought to be lethal. Hence, prenatal ultrasound detection of bilateral radial ray anomalies with present thumbs376 necessitates not only CNV interrogation for the chromosome 1q21.1 deletion, but also RBM8A gene sequencing to establish the molecular diagnosis of TAR syndrome.377, 378

CHAPTER 14

Determination of the pathogenic variants in a family will allow additional carrier screening and the opportunity for future PGT and prenatal diagnosis. There is a paucity of reports on the prenatal diagnosis of SCA.379–381 Currently there are at least 40 types of SCA, and a number associated with other neurological findings such as deafness. In a Cuban study of patients with SCA type 2, 28 out of 51 known presymptomatic carriers had affected fetuses, and 20 (71.43 percent) elected to terminate their pregnancies.380 These authors subsequently have drawn attention to couples where one has an expanded and a large normal allele, which may result in occurrences greater than 50 percent.381 Earlier, authors in Taiwan drew attention to the prenatal diagnosis of SCA3, in a case in which there was a contraction of repeat expansions from 78 in the father to 74 in the fetus.379 In another study of SCA patients examining the perceptions regarding reproductive options, the majority were most interested in PGT to avoid having affected children.382 A special caveat exists when performing prenatal diagnosis for repeat expansion disorders. Not infrequently, a fetus will appear to be homozygous for a normal allele (e.g. SCA2). It is important to know the allele sizes of the affected parent (and often the unaffected parent) in order not to miss a very large fetal expansion. Use of repeat primed PCR assay provides added reassurance of a normal-size homozygous allele.383, 384 Major advances have occurred in the understanding of the neurogenetic basis of epilepsy. Scores of newly recognized genes and their causal mutations have been documented, representing primarily de novo autosomal dominant genes, but also autosomal recessive and X-linked ones.385–389 Molecular diagnosis of those individuals with syndromic and nonsyndromic epilepsy with or without intellectual disability has improved anticipatory guidance for those affected, allowed more appropriate targeted therapies, and the ability to recognize family members who may be mildly affected.390, 391 A recent study of patients with epilepsy and intellectual disability has demonstrated an additional 10 percent positive molecular diagnosis if the previously reported negative WES data was reanalyzed 6–12 months later.392 Hence, as the databases and literature are updated, so too should our previously negative data be

Molecular Genetics and Prenatal Diagnosis 589

reanalyzed, especially in those in whom a VUS has been reported.393, 394 Additional molecular approaches including high-throughput automated patch clamping to assess the functional properties of a detected VUS have proven to be beneficial in the classification of these variants in the SCN5A gene.395 A parent with a history of seizures needs careful clinical assessment as gene analysis would be available and prenatal diagnosis and PGT would be an option for such conditions as tuberous sclerosis, neurofibromatosis, incontinentia pigmenti, and any gene in which the pathogenic variant(s) are established. The genetic basis of many myopathies has been resolved providing opportunities for prenatal diagnosis and PGT after mutation detection. Myotonic dystrophies (DM) types 1 and 2 as well as DMD/Becker muscular dystrophy were discussed in Chapter 1. A major concern is the affected pregnant woman with DM1 giving birth to a child with the severe congenital DM1, given the high risk of anticipation. A contrasting concern for DMD/Becker muscular dystrophy “noncarrier” females who have had one affected son is the 4–8 percent risk of recurrence due to gonadal mosaicism.98 Mutation detection in the proband is therefore crucial prior to further childbearing. Rarely two deletions or two duplications may be found in the DMD gene even if one such change did not disrupt the reading frame.396 A large study of 1,053 Chinese patients with DMD/Becker muscular dystrophy noted large rearrangements in the dystrophin gene in 70.56 percent of probands, 59.35 percent and 11.21 percent having deletions or duplications, respectively.397 A salutary lesson has emerged from a Dutch study of DMD during the periods 1961–1974 and 1993–2002.98 Remarkably the incidence of DMD did not decline, with the percentage of first affected boys increasing from 62 percent to 88 percent. The authors concluded that it took up to 5 years for a first diagnosis of DMD. Consequently recurrence was common. Moreover, female fetuses were not tested for their carrier status and years later went on to have affected children. They pressed for a change in policy with special reference to determining female carrier status during prenatal diagnosis and introducing neonatal screening of males. Requests for prenatal diagnosis of severe skin disorders are uncommon. Originally fetal skin

590 Genetic Disorders and the Fetus

biopsy was the only means to achieve prenatal detection. Subsequent recognition of causative genes has enabled molecular prenatal diagnosis. Consequently, Harlequin ichthyosis has been detected prenatally via sequencing of the ABCA12 gene.398 Prenatal sonographic features have been described.399 Despite familial phenotypic variability, prenatal diagnosis has also been successfully achieved or the diagnosis excluded by sequencing of the TGM1 gene in families with lamellar ichthyosis.400, 401 Prenatal diagnosis of epidermolytic ichthyosis or hyperkeratosis has so far been achieved by molecular analysis of ABCA12. In fact, for all three clinical phenotypes mentioned, mutations in this gene represent a major cause of severe autosomal recessive congenital ichthyoses.402, 403 X-linked steroid sulfatase deficiency (STS), most often suspected by the finding of low maternal serum estriol in the second trimester of pregnancy, is usually determined prenatally by demonstration of a deletion in the STS gene present in about 90 percent of families.404, 405 In about 10 percent of families, however, a contiguous gene deletion syndrome may be present and include the association of Kallman syndrome and chondrodysplasia punctata.406 Larger deletions in the STS gene are likely to be associated with intellectual disability and hypogonadism. Prenatal diagnosis of steroid sulfatase deficiency by FISH or microarray has been made repeatedly.407, 408 Cases of epidermolysis bullosa come to attention for prenatal diagnosis either because an unexplained elevation of amniotic fluid 𝛼-fetoprotein is observed or following the birth of a previously affected child. PGT has also been achieved with exclusion of the disorder by haplotyping in a fetus with Herlitz junctional epidermolysis bullosa.409 There are four categories and multiple subcategories of the autosomal recessive epidermolysis bullosa. For many years, fetal skin biopsy was used for prenatal diagnosis of lethal epidermolysis bullosa subtypes. This has now been replaced by NGS approaches.410 The prenatal detection of adult-onset potentially lethal disorders has steadily gained traction, including the neurodegenerative disorders, cardiomyopathies, and malignancies. The prenatal detection of a retinoblastoma, initially by linkage analysis and subsequently by routine molecular techniques, has been available for decades.

DNA analysis detects about 92 percent of the RB1 gene mutations in either bilateral and/or familial cases.411 An important potential pitfall is the presence of mosaicism.412, 413 Incidental detection of a fetal chromosomal abnormality involving the retinoblastoma gene locus on 13p following detection, for example, of increased nuchal translucency, could infer a likely future retinoblastoma.414 Although first- or second-trimester prenatal diagnosis for breast/ovarian cancer gene mutations are available, a likely more common future choice is PGT.415, 416 Similarly, patients at risk of having offspring with familial adenomatous polyposis, hereditary nonpolyposis colon cancer, or multiple endocrine neoplasia may select PGT or prenatal diagnosis.417–420 Noninvasive prenatal screening for these disorders may be available with known mutations (see Chapter 8).

Prenatal diagnosis of mitochondrial disorders Mitochondrial disorders are more common than originally thought and have an estimated prevalence of at least one case in 7,500 individuals.421, 422 Mitochondrial disorders may be due to mutations in either mtDNA (∼ 1/3) or nuclear DNA (∼ 2/3).423, 424 Primary mitochondrial diseases are due to defects in mtDNA, which are transmitted via maternal inheritance. mtDNA is a double-stranded circular molecule with 37 genes. This means that all mitochondria in the zygote derive from the ovum, thus far with rare exceptions.425–427 Hence, an affected mother with a mitochondrial disorder would transmit to all of her children, but subsequently her sons, while affected, would not be transmitters. A characteristic feature of this form of inheritance is that the pathogenic mutations in mtDNA are present in some but not necessarily all mitochondria. This situation, known as heteroplasmy, will be highly variable with greater clinical manifestations, reflecting high mutant loads. Moreover, the highly variable distribution of mutations within mtDNA will vary from tissue to tissue. Indeed, heteroplasmy may also occur in single mitochondria in which both normal and mutated mtDNA can be found. Tissues that are highly dependent on oxidative metabolism, such as the brain, heart, skeletal muscle, retina, renal

CHAPTER 14

tubules, and endocrine glands, are particularly vulnerable to the effects of mutations in mtDNA. Prenatal diagnosis of mitochondrial disorders is fraught with challenges. Extremely careful genetic counseling will inform an affected mother of the likely 100 percent transmission of her mutation to all her offspring. Key to the clinical manifestations is the size of the mutant load transmitted and the tissue distribution of the abnormal mitochondria. Hence, at-risk parents will have to understand that assessment of the mutant load from chorionic villi or amniocyte cells may not necessarily reflect ultimate fetal health and welfare.428 A low mutant load may not necessarily result in a child with few or no clinical features and, moreover, may be uninformative about future health. Vachin et al. have shown more reliable mutant load determinations from amniocytes than from chorionic villi.429 Nesbitt et al. describe their experience with prenatal diagnosis of mitochondrial disorders in the United Kingdom.430 Their approach includes ascertaining the degree of maternal heteroplasmy (blood and urine) and that of affected and unaffected maternal relatives. Utilization of NGS as well as quantitative real-time PCR assays have improved the accurate detection of heteroplasmy in the tissue being studied.431–433 Notwithstanding the obvious lack of guarantees in these circumstances, a number of cases have been reported for the prenatal diagnosis of Leigh syndrome, more specifically of the m.T8993G mutation434–437 and the m.T8993C mutation.438 “Successful” prenatal diagnosis of MELAS (myopathy, encephalopathy, lactic acidosis and stroke-like syndrome) has been reported.439 As some of the mitochondrial disorders, especially with significant heteroplasmy, have symptomatic presentation in adulthood, caution is needed with any prenatal prognostications. Prenatal diagnosis of nuclear mitochondrial genes transmitted as monogenic disorders is feasible if the familial mutations are known. Similarly, PGT is available for nuclear mitochondrial genes with known mutations (see Chapter 2). Amato et al. described novel three-parent in vitro fertilization (IVF) techniques designed to avoid the maternal transmission of mitochondrial disorders.440 Maternal spindle transfer is an approach in the setting of IVF to prevent the transmission of mtDNA mutations.441–443 Further larger studies are needed to determine efficacy and success rates.

Molecular Genetics and Prenatal Diagnosis 591

Reporting incidental (secondary) results SNP microarray analysis, WES, and WGS have inevitably led to the recognition of genomic abnormalities that were not the purpose of the intended study. When such unexpected observations reveal mutations that enable prediction with significant life-threatening risk, a responsibility inures to communicate with the patient or family. The American College of Medical Genetics and Genomics (ACMG) has published recommendations for reporting incidental findings444 and provided a “minimum” list of 56 (now 59) serious monogenic disorders requiring communication445, 446 (Table 14.5). This list, to which other disorders will undoubtedly be added, focuses on conditions where treatment is available or surveillance is necessary. Not included in the list are structural variants (e.g. translocations and inversions), repeat expansions, or CNVs. The ACMG report emphasized that a negative incidental findings report should not be misconstrued to imply the absence of a pathogenic variant. Moreover, the ACMG weighed the practical issues and limitations of genetic counseling in clinical practice as well as patients’ autonomy. A fiduciary duty to prevent harm by warning patients was recognized for both clinicians and reporting laboratories. Informed consent for patients undergoing sequencing about possible incidental findings provides them with the choice to opt out. A recent Dutch study examining 1,640 anonymized healthy individuals by WES, identified 1 in 38 with a medically actionable dominant (likely) pathogenic variant in the 59 ACMG recommended genes to be reported.447 Additional studies of varying sizes examined medically actionable pathogenic variants found in 56–114 genes and reported less than 3.4 percent.116, 448, 449 These results vary depending on gene selection, population studied, and criteria utilized to determine variant pathogenicity. Studies regarding patients’ perspectives,450 methods for return of the findings,451 as well as management,452 and implications for genetic counseling, are germane to prenatal diagnosis.68 Policies not to perform presymptomatic or predictive genetic testing of children have been in place

592 Genetic Disorders and the Fetus

Table 14.5 Conditions, genes, and variants recommended for return of incidental findings in clinical sequencing Phenotype Hereditary breast and ovarian cancer

Gene

Inheritancea

Variants to reportb

BRCA1

AD

KP and EP KP and EP

BRCA2 Li–Fraumeni syndrome

TP53

AD

Peutz–Jeghers syndrome

STK11

AD

KP and EP

Lynch syndrome

MLH1

AD

KP and EP

MSH2 MSH6 PMS2 Familial adenomatous polyposis

APC

AD

KP and EP

MYH-associated polyposis; adenomas,

MUTYH

ARc

KP and EP

multiple colorectal, FAP type 2; colorectal adenomatous polyposis, autosomal recessive, with pilomatricomas Juvenile polyposis

BMPR1A SMAD4

AD

KP and EP

Von Hippel–Lindau syndrome

VHL

AD

KP and EP

Multiple endocrine neoplasia type 1

MEN1

AD

KP and EP

Multiple endocrine neoplasia type 2

RET

AD

KP

Familial medullary thyroid cancer (FMTC)

RET

AD

KP

PTEN hamartoma tumor syndrome

PTEN

AD

KP and EP

Retinoblastoma

RB1

AD

KP and EP

Hereditary

SDHD

AD

KP and EP

paraganglioma–pheochromocytoma

SDHAF2

KP

syndrome

SDHC

KP and EP

SDHB Tuberous sclerosis complex

TSC1

KP and EP AD

KP and EP

TSC2 WT1-related Wilms tumor

WT1

AD

KP and EP

Neurofibromatosis type 2

NF2

AD

KP and EP

Ehlers–Danlos syndrome – vascular type

COL3A1

AD

KP and EP

Marfan syndrome, Loeys–Dietz syndromes,

FBN1

AD

KP and EP

and familial thoracic aortic aneurysms

TGFBR1

and dissections

TGFBR2 SMAD3 ACTA2 MYH11

Hypertrophic cardiomyopathy, dilated cardiomyopathy

MYBPC3

AD

KP and EP

MYH7

KP

TNNT2

KP and EP

TNNI3

KP

TPM1 MYL3 ACTC1 PRKAG2 GLA

XL

KP and EP (hemi, het, hom)

MYL2

AD

KP and EP

AD

KP

LMNA Catecholaminergic polymorphic ventricular

RYR2

tachycardia (Continued)

CHAPTER 14

Molecular Genetics and Prenatal Diagnosis 593

Table 14.5 (Continued) Phenotype

Gene

Inheritancea

Arrhythmogenic right ventricular

PKP2

AD

cardiomyopathy

Variants to reportb KP and EP

DSP

KP

DSC2

KP and EP

TMEM43 DSG2 Romano–Ward long QT syndromes types 1, 2, and 3, Brugada syndrome

KCNQ1

AD

KP and EP

KCNH2 SCN5A

Familial hypercholesterolemia

LDLR

SD

KP and EP

APOB

SD

KP

PCSK9

AD

Wilson disease

ATP7B

ARc

KP and EP

Ornithine transcarbamylase deficiency

OTC

XL

KP and EP (hemi, het, hom)

Malignant hyperthermia susceptibility

RYR1

AD

KP

CACNA1S a Some

conditions that may demonstrate semidominant inheritance (SD) have been indicated as autosomal dominant

(AD) for the sake of simplicity. Others have been labeled as X-linked (XL). b KP:

known pathogenic, sequence variation is previously reported and is a recognized cause of the disorder; EP: expected

pathogenic, sequence variation is previously unreported and is of the type which is expected to cause the disorder. The recommendation not to report expected pathogenic variants for some genes is due to the recognition that truncating variants, the primary type of expected pathogenic variants, are not an established cause of some diseases on the list. c We

recommend searching only for individuals with biallelic mutations.

Source: Modified from Kalia SS, Adelman K, Bale SJ, et al. Recommendations for reporting of secondary findings in clinical exome and genome sequencing, 2016 update (ACMG SF v2.0): a policy statement of the American College of Medical Genetics and Genomics [published correction appears in Genet Med. 2017 Apr;19:484]. Genet Med 2017;19:249.446

for at least two decades.453–455 In the face of discovery of an incidental finding that may have serious health implications for a parent or concerning future reproduction, the age bar for testing has been lifted.

Ethical considerations in prenatal testing As our molecular and bioinformatic technologies rapidly progress, a crucial element in providing

prenatal diagnostic genomics is the recognition and understanding of parental perceptions and ethical implications (see Chapter 37 for in-depth discussion). Studies that examine parental, clinician, and researchers’ perspectives on prenatal genomics can promote more optimal care.456–459 The essential discussion of the ethical and counseling challenges of prenatal genomics will hopefully continue to positively impact the provision of standard prenatal care.460–463

References 1. Crawford K, Bracher-Smith M, Owen D, et al. Medical

3. Baldridge D, Spillmann RC, Wegner DJ, et al. Pheno-

consequences of pathogenic CNVs in adults: analysis

typic expansion of KMT2D-related disorder: Beyond

of the UK Biobank. J Med Genet 2019;56:131.

Kabuki syndrome. Am J Med Genet A 2020;182:1053.

2. Scott TM, Guo H, Eichler EE, et al. BAZ2B haploinsuf-

4. Ranganath P, Perala S, Nair L, et al. A newly recognized

ficiency as a cause of developmental delay, intellectual

multiple malformation syndrome with caudal regres-

disability, and autism spectrum disorder. Hum Mutat 2020;41:921.

sion associated with a biallelic c.402G>A variant in TBX4. Eur J Hum Genet 2020;28:669.

594 Genetic Disorders and the Fetus

5. Jurinke C, Oeth P, van den Boom D. MALDI-TOF mass spectrometry: a versatile tool for high-performance DNA analysis. Mol Biotechnol 2004;26:147. 6. Arcila M, Lau C, Nafa K, et al. Detection of KRAS and BRAF mutations in colorectal carcinoma roles for high-sensitivity locked nucleic acid-PCR sequencing and broad-spectrum mass spectrometry genotyping. J Mol Diagn 2011;13:64. 7. Salk JJ, Schmitt MW, Loeb LA. Enhancing the accuracy of next-generation sequencing for detecting rare and subclonal mutations. Nat Rev Genet 2018;19:269. 8. Tong Y, Shen S, Jiang H, et al. Application of digital PCR in detecting human diseases associated gene mutation. Cell Physiol Biochem 2017;43:1718. 9. Southern E. Detection of specific sequences among DNA fragments separated by gel electrophoresis. J Mol Biol 1975;98:503. 10. Tanaka E, Maruyama H, Morino H, et al. Detection of large expansions in SCA8 using a fluorescent repeat-primed PCR assay. Hiroshima J Med Sci 2011;60:63. 11. Singh S, Zhang A, Dlouhy S, et al. Detection of large expansions in myotonic dystrophy type 1 using triplet primed PCR. Front Genet 2014;5:94. 12. Orpana AK, Ho TH, Alagrund K, et al. Novel heat pulse extension-PCR-based method for detection of large CTG-repeat expansions in myotonic dystrophy type 1. J Mol Diagn 2013;15:110. 13. Suh E, Grando K, Van Deerlin VM. Validation of a long-read PCR assay for sensitive detection and sizing of C9orf72 hexanucleotide repeat expansions. J Mol Diagn 2018;20:871. 14. Milunsky JM, Maher TA, Loose BA, et al. XL PCR for the detection of large trinucleotide expansions in juvenile Huntington’s disease. Clin Genet 2003;64:70. 15. Sanger F, Coulson A. A rapid method for determining sequences in DNA by primed synthesis with DNA polymerase. J Mol Biol 1975;94:441. 16. Tyburczy ME, Dies KA, Glass J, et al. Mosaic and intronic mutations in TSC1/TSC2 explain the majority of TSC patients with no mutation identified by conventional testing. PLoS Genet 2015;11:e1005637. 17. Arnold AM, Morak M, Benet-Pagès A, et al. Targeted deep-intronic sequencing in a cohort of unexplained cases of suspected Lynch syndrome. Eur J Hum Genet 2020;28:597. 18. Schouten JP, McElgunn CJ, Waaijer R, et al. Relative quantification of 40 nucleic acid sequences bymultiplex ligation-dependent probe amplification. Nucleic Acids Res 2002;30:e57.

19. Milunsky JM, Maher TA, Ito M, et al. The value of MLPA in Waardenburg syndrome. Genet Test 2007;11:179. 20. Kozlowski P, Roberts P, Dabora S, et al. Identification of 54 large deletions/duplications in TSC1 and TSC2 using MLPA, and genotype–phenotype correlations. Hum Genet 2007;121:389. 21. Moelans CB, Atanesyan L, Savola SP, et al. Methylation-specific multiplex ligation-dependent probe amplification (MS-MLPA). Methods Mol Biol 2018;1708:537. 22. Schouten J, van Vught P, Galjaard RJ. Multiplex ligation-dependent probe amplification (MLPA) for prenatal diagnosis of common aneuploidies. Methods Mol Biol 2019;1885:161. 23. Mei R, Galipeau PC, Prass C, et al. Genome-wide detection of allelic imbalance using human SNPs and high density DNA arrays. Genome Res 2000;10:1126. 24. Schuster SC. Next-generation sequencing transforms today’s biology. Nat Methods 2008;5:16. 25. Bentley DR, Balasubramanian S, Swerdlow HP, et al. Accurate whole human genome sequencing using reversible terminator chemistry. Nature 2008;456:53. 26. Downie L, Halliday J, Burt R, et al. Exome sequencing in infants with congenital hearing impairment: a population-based cohort study. Eur J Hum Genet 2020;28:587. 27. Jiman OA, Taylor RL, Lenassi E, et al. Diagnostic yield of panel-based genetic testing in syndromic inherited retinal disease. Eur J Hum Genet 2020;28:576. 28. Hansen MC, Haferlach T, Nyvold CG. A decade with whole exome sequencing in haematology. Br J Haematol 2020;188:367. 29. Adams DR, Eng CM. Next-generation sequencing to diagnose suspected genetic disorders. N Engl J Med 2019;380:201. 30. Wise AL, Manolio TA, Mensah GA, et al. Genomic medicine

for

undiagnosed

diseases.

Lancet

2019;394:533. 31. Farwell KD, Shahmirzadi L, El-Khechen D, et al. Enhanced utility of family-centered diagnostic exome sequencing with inheritance model-based analysis: results from 500 unselected families with undiagnosed genetic conditions. Genet Med 2015;17:578. 32. Greer SU, Ji HP. Structural variant analysis for linked-read sequencing data with gemtools. Bioinformatics 2019;35:4397. 33. Ngo KJ, Rexach JE, Lee H, et al. A diagnostic ceiling for exome sequencing in cerebellar ataxia and related neurological disorders. Hum Mutat 2020;41:487.

CHAPTER 14

34. Retterer K, Juusola J, Cho MT, et al. Clinical application of whole-exome sequencing across clinical indications. Genet Med 2016;18:696. 35. Tan TY, Dillon OJ, Stark Z, et al. Diagnostic impact and cost-effectiveness of whole-exome sequencing for ambulant children with suspected monogenic conditions. JAMA Pediatr 2017;171:855. 36. Alfares A, Aloraini T, Subaie LA, et al. Whole-genome sequencing offers additional but limited clinical utility compared with reanalysis of whole-exome sequencing. Genet Med 2018;20:1328. 37. Elliott AM, du Souich C, Lehman A, et al. RAPIDOMICS: rapid genome-wide sequencing in a neonatal intensive care unit-successes and challenges. Eur J Pediatr 2019;178:1207. 38. Borghesi A, Mencarelli MA, Memo L, et al. Intersociety policy statement on the use of whole-exome sequencing in the critically ill newborn infant. Ital J Pediatr 2017;43:100. 39. Ceyhan-Birsoy O, Machini K, Lebo MS, et al. A curated gene list for reporting results of newborn genomic sequencing. Genet Med 2017;19:809. 40. Kuehne B, Heine E, Dafsari HS, et al. Use of whole exome sequencing in the NICU: Case of an extremely low birth weight infant with syndromic features. Mol Cell Probes 2019;45:89. 41. Leone MP, Palumbo P, Palumbo O, et al. The recurrent SETBP1 c.2608G > A, p.(Gly870Ser) variant in a patient with Schinzel-Giedion syndrome: an illustrative case of the utility of whole exome sequencing in a critically ill neonate. Ital J Pediatr 2020;46:74. 42. Maxmen A. Exome sequencing deciphers rare diseases. Cell 2011;144:635. 43. Yang Y, Muzny DM, Reid JG, et al. Clinical whole-exome sequencing for the diagnosis of mendelian disorders. N Engl J Med 2013;369:1502. 44. Han JY, Lee IG. Genetic tests by next-generation sequencing in children with developmental delay and/or intellectual disability. Clin Exp Pediatr 2020;63:195. 45. Stark Z, Tan TY, Chong B, et al. A prospective evaluation of whole-exome sequencing as a first-tier molecular test in infants with suspected monogenic disorders. Genet Med 2016;18:1090. 46. Sánchez Fernández I, Loddenkemper T, Gaínza-Lein M, et al. Diagnostic yield of genetic tests in epilepsy: a meta-analysis and cost-effectiveness study [published online ahead of print, 2019 Jan 4]. Neurology 2019;92:e418. 47. Mellis R, Chandler N, Chitty LS. Next-generation sequencing and the impact on prenatal diagnosis. Expert Rev Mol Diagn 2018;18:689.

Molecular Genetics and Prenatal Diagnosis 595

48. Vora NL, Hui L. Next-generation sequencing and prenatal ‘omics: advanced diagnostics and new insights into human development. Genet Med 2018;20:791. 49. Normand EA, Alaimo JT, Van den Veyver IB. Exome and genome sequencing in reproductive medicine. Fertil Steril 2018;109:213. 50. de Koning MA, Haak MC, Adama van Scheltema PN, et al. From diagnostic yield to clinical impact: a pilot study on the implementation of prenatal exome sequencing in routine care. Genet Med 2019;21:2303. 51. Liu Y, Wang L, Yang YK, et al. Prenatal diagnosis of fetal skeletal dysplasia using targeted next-generation sequencing: an analysis of 30 cases. Diagn Pathol 2019;14:76. 52. Han J, Yang YD, He Y, et al. Rapid prenatal diagnosis of skeletal dysplasia using medical trio exome sequencing: benefit for prenatal counseling and pregnancy management. Prenat Diagn 2020;40:577. 53. Petrovski S, Aggarwal V, Giordano JL, et al. Whole-exome sequencing in the evaluation of fetal structural anomalies: a prospective cohort study. Lancet 2019;393:758. 54. Lord J, McMullan DJ, Eberhardt RY, et al. Prenatal exome sequencing analysis in fetal structural anomalies detected by ultrasonography (PAGE): a cohort study. Lancet 2019;393:747. 55. Ferretti L, Mellis R, Chitty LS. Update on the use of exome sequencing in the diagnosis of fetal abnormalities. Eur J Med Genet 2019;62:103663. 56. Felice V, Abhyankar A, Jobanputra V. Prenatal diagnosis by whole exome sequencing in fetuses with ultrasound abnormalities. Methods Mol Biol 2019;1885:267. 57. Leung GKC, Mak CCY, Fung JLF, et al. Identifying the genetic causes for prenatally diagnosed structural congenital anomalies (SCAs) by whole-exome sequencing (WES). BMC Med Genomics 2018;11:93. 58. Aarabi M, Sniezek O, Jiang H, et al. Importance of complete phenotyping in prenatal whole exome sequencing. Hum Genet 2018;137:175. 59. Mone F, Quinlan-Jones E, Kilby MD. Clinical utility of exome sequencing in the prenatal diagnosis of congenital anomalies: a review. Eur J Obstet Gynecol Reprod Biol 2018;231:19. 60. Lei TY, Fu F, Li R, et al. Whole-exome sequencing for prenatal diagnosis of fetuses with congenital anomalies of the kidney and urinary tract. Nephrol Dial Transplant 2017;32:1665. 61. Traversa A, Bernardo S, Paiardini A, et al. Prenatal whole exome sequencing detects a new homozygous

596 Genetic Disorders and the Fetus

62.

63.

64.

65.

66.

67.

68.

69.

70.

71.

72.

73.

74.

fukutin (FKTN) mutation in a fetus with an ultrasound suspicion of familial Dandy-Walker malformation. Mol Genet Genomic Med 2020;8:e1054. Linpeng S, Liu J, Pan J, et al. Diagnosis of Joubert syndrome 10 in a fetus with suspected Dandy-Walker variant by WES: a novel splicing mutation in OFD1. Biomed Res Int 2018;2018:4032543. Yates CL, Monaghan KG, Copenheaver D, et al. Whole-exome sequencing on deceased fetuses with ultrasound anomalies: expanding our knowledge of genetic disease during fetal development. Genet Med 2017;19:1171. Stals KL, Wakeling M, Baptista J, et al. Diagnosis of lethal or prenatal-onset autosomal recessive disorders by parental exome sequencing. Prenat Diagn 2018;38:33. Wou K, Chung WK, Wapner RJ. Laboratory considerations for prenatal genetic testing. Semin Perinatol 2018;42:307. Vears DF, Niemiec E, Howard HC, et al. Analysis of VUS reporting, variant reinterpretation and recontact policies in clinical genomic sequencing consent forms. Eur J Hum Genet 2018;26:1743. Abou Tayoun AN, Spinner NB, Rehm HL, et al. Prenatal DNA sequencing: clinical, counseling, and diagnostic laboratory considerations. Prenat Diagn 2018;38:26. Best S, Wou K, Vora N, et al. Promises, pitfalls and practicalities of prenatal whole exome sequencing. Prenat Diagn 2018;38:10. Linderman MD, Brandt T, Edelmann L, et al. Analytical validation of whole exome and whole genome sequencing for clinical applications. BMC Med Genomics 2014;7:20. Seneca S, Vancampenhout K, Van Coster R, et al. Analysis of the whole mitochondrial genome: translation of the Ion Torrent Personal Genome Machine system to the diagnostic bench. Eur J Hum Genet 2014; 23:41. Caspar SM, Dubacher N, Kopps AM, et al. Clinical sequencing: From raw data to diagnosis with lifetime value. Clin Genet 2018;93:508. Vakkilainen S, Skoog T, Einarsdottir E, et al. The human long non-coding RNA gene RMRP has pleiotropic effects and regulates cell-cycle progression at G2. Sci Rep 2019;9:13758. Gotway G, Crossley E, Kozlitina J, et al. Clinical exome studies have inconsistent coverage. Clin Chem 2020;66:199. Mickle J, Milunsky A, Amos JA, et al. Congenital unilateral absence of the vas deferens: a heterogeneous disorder with two distinct subpopulations based upon aetiology and mutational status of the cystic fibrosis gene. Hum Reprod 1995;10:1728.

75. Sebastiani P, Solovieff N, Hartley SW, et al. Genetic modifiers of the severity of sickle cell anemia identified through a genome-wide association study. Am J Hematol 2010;85:29. 76. Wilhelm BT, Landry JR. RNA-Seq-quantitative measurement of expression through massively parallel RNA-sequencing. Methods 2009;48:249. 77. Bibikova M, Fan JB. Genome-wide DNA methylation profiling. Wiley Interdiscip Rev Syst Biol Med 2010;2:210. 78. Dor Y, Cedar H. Principles of DNA methylation and their implications for biology and medicine. Lancet 2018;392:777. 79. Stavropoulos DJ, Merico D, Jobling R, et al. Whole genome sequencing expands diagnostic utility and improves clinical management in pediatric medicine. NPJ Genom Med 2016;1:15012. 80. Mao Q, Chin R, Xie W, et al. Advanced whole-genome sequencing and analysis of fetal genomes from amniotic fluid. Clin Chem 2018;64:715. 81. Zhu X, Hu Y, Li J. [Interpretation of the joint position statement of the International Prenatal Diagnosis Society, the Maternal and Fetal Medicine Association and the Perinatal Quality Foundation on the use of whole genome sequencing for fetal diagnosis]. Zhonghua Fu Chan Ke Za Zhi [Chinese J Obstetr Gynecol] 2019;54:645. 82. Richards CS, Bale S, Bellissimo DB, et al. ACMG recommendations for standards for interpretation and reporting of sequence variations: revisions 2007. Genet Med 2008;10:294. 83. Harrison SM, Biesecker LG, Rehm HL. Overview of specifications to the ACMG/AMP variant interpretation guidelines. Curr Protoc Hum Genet 2019;103:e93. 84. Krsti´c N, Obiˇcan SG. Current landscape of prenatal genetic screening and testing. Birth Defects Res 2020;112:321. 85. Bell CJ, Dinwiddie DL, Miller NA, et al. Carrier testing for severe childhood recessive diseases by next generation sequencing. Sci Transl Med 2011;3:65ra4. 86. Nazareth SB, Lazarin GA, Goldberg JD. Changing trends in carrier screening for genetic disease in the United States. Prenat Diagn 2015;35:931. 87. Guo MH, Gregg AR. Estimating yields of prenatal carrier screening and implications for design of expanded carrier screening panels. Genet Med 2019;21:1940. 88. Ben-Shachar R, Svenson A, Goldberg JD, et al. A data-driven evaluation of the size and content of expanded carrier screening panels. Genet Med 2019;21:1931.

CHAPTER 14

89. Metcalfe SA. Genetic counselling, patient education, and informed decision-making in the genomic era. Semin Fetal Neonatal Med 2018;23:142. 90. Gregg AR. Expanded carrier screening. Obstet Gynecol Clin North Am 2018;45:103. 91. Gregg AR, Edwards JG. Prenatal genetic carrier screening in the genomic age. Semin Perinatol 2018;42:303. 92. Romão EA, Lourenço CM, Júnior WM, et al. What lies beneath: Fabry nephropathy in a female patient with severe cerebrovascular disease. Clin Nephrol 2013;79:330. 93. Prior TW. Professional Practice and Guidelines Committee. Carrier screening for spinal muscular atrophy. Genet Med 2008;10:840. 94. Su YN, Hung CC, Lin SY, et al. Carrier screening for spinal muscular atrophy (SMA) in 107,611 pregnant women during the period 2005–2009: a prospective population-based cohort study. PLoS One 2011;6:e17067. 95. Xu Y, Xiao B, Liu Y, et al. Identification of novel SMN1 subtle mutations using an allelic-specific RT-PCR. Neuromuscul Disord 2020;30:219. 96. Luo M, Liu L, Peter I, et al. An Ashkenazi Jewish SMN1 haplotype specific to duplication alleles improves panethnic carrier screening for spinal muscular dystrophy. Genet Med 2014;16:149. 97. Azad AK, Huang CK, Jin H, et al. Enhanced carrier screening for spinal muscular atrophy: detection of silent (SMN1: 2 + 0) carriers utilizing a novel TaqMan genotyping method [published online ahead of print, 2019 Dec 26]. Lab Med 2019;lmz088. 98. Helderman-van den Enden AT, van den Bergen JC, Breuning MH, et al. Duchenne/Becker muscular dystrophy in the family: have potential carriers been tested at a molecular level? Clin Genet 2011;79:236. 99. Fraser HG, Redmond RZ, Scotcher DF. Experiences of women who have had carrier testing for Duchenne muscular dystrophy and Becker muscular dystrophy during adolescence. J Genet Couns 2018;27:1349. 100. Makrythanasis P, Nelis M, Santoni FA, et al. Diagnostic exome sequencing to elucidate the genetic basis of likely recessive disorders in consanguineous families. Hum Mutat 2014;35:1203. 101. Eaton A, Hartley T, Kernohan K, et al. When to think outside the autozygome: Best practices for exome sequencing in “consanguineous” families. Clin Genet 2020;97:835. 102. Gezdirici A, Ekiz A, Güleç EY, et al. How necessary is to analyze PTPN11 gene in fetuses with first trimester cystic hygroma and normal karyotype?. J Matern Fetal Neonatal Med 2017;30:938.

Molecular Genetics and Prenatal Diagnosis 597

103. Schreurs L, Lannoo L, De Catte L, et al. First trimester cystic hygroma colli: Retrospective analysis in a tertiary center. Eur J Obstet Gynecol Reprod Biol 2018;231:60. 104. Bezniakow N, Gos M, Obersztyn E. The RASopathies as an example of RAS/MAPK pathway disturbances – clinical presentation and molecular pathogenesis of selected syndromes. Dev Period Med 2014;18:285. 105. Liao J, Mehta L. Molecular genetics of Noonan syndrome and RASopathies. Pediatr Endocrinol Rev 2019;16(Suppl 2):435. 106. Rauen KA. The RASopathies. Annu Rev Genomics Hum Genet 2013;14:355. 107. Aoki Y, Niihori T, Inoue S, et al. Recent advances in RASopathies. J Hum Genet 2016;61:33. 108. Tidyman WE, Rauen KA. Expansion of the RASopathies. Curr Genet Med Rep 2016;4:57. 109. Stuurman KE, Joosten M, van der Burgt I, et al. Prenatal ultrasound findings of rasopathies in a cohort of 424 fetuses: update on genetic testing in the NGS era. J Med Genet 2019;56:654. 110. Redin C, Gérard B, Lauer J, et al. Efficient strategy for the molecular diagnosis of intellectual disability using targeted high-throughput sequencing. J Med Genet 2014;51:724. 111. Shkedi-Rafid S, Dheensa S, Crawford G, et al. Defining and managing incidental findings in genetic and genomic practice. BMJ 2014;51:715. 112. Pangalos C, Hagnefelt B, Lilakos K, et al. First applications of a targeted exome sequencing approach in fetuses with ultrasound abnormalities reveals an important fraction of cases with associated gene defects. PeerJ 2016;4:e1955. 113. MacArthur DG, Balasubramanian S, Frankish A, et al. A systematic survey of loss-of-function variants in human protein-coding genes. Science 2012;335:823. 114. MacArthur DG, Tyler-Smith C. Loss-of-function variants in the genomes of healthy humans. Hum Mol Genet 2010;19:R125 115. Cooper GM, Shendure J. Needles in stacks of needles: finding disease-causal variants in a wealth of genomic data. Nat Rev Genet 2011;12:628. 116. Dorschner MO, Amendola LM, Turner EH, et al. Actionable, pathogenic incidental findings in 1,000 participants’ exomes. Am J Hum Genet 2013;93:631. 117. Lawrence L, Sincan M, Markello T, et al. The implications of familial incidental findings from exome sequencing: the NIH undiagnosed diseases program experience. Genet Med 2014;16:741. 118. Meric-Bernstam F, Brusco L, Daniels M, et al. Incidental germline variants in 1000 advanced cancers on a

598 Genetic Disorders and the Fetus

119.

120.

121.

122.

123.

124.

125. 126.

127.

128.

129.

130.

131.

132.

prospective somatic genomic profiling protocol. Ann Oncol 2016;27:795. La Spada AR, Taylor JP. Repeat expansion disease: progress and puzzles in disease pathogenesis. Nat Rev Genet 2010;1:247. López Castel A, Cleary JD, Pearson CE. Repeat instability as the basis for human diseases and as a potential target for therapy. Nat Rev Mol Cell Biol 2010;11:165. Mootha VV, Gong X, Ku HC, et al. Association and familial segregation of CTG18.1 trinucleotide repeat expansion of TCF4 gene in Fuchs’ endothelial corneal dystrophy. Invest Ophthalmol Vis Sci 2014;55:33. Rodriguez CM, Todd PK. New pathologic mechanisms in nucleotide repeat expansion disorders. Neurobiol Dis 2019;130:104515. Tian Y, Wang JL, Huang W, et al. Expansion of human-specific GGC repeat in neuronal intranuclear inclusion disease-related disorders. Am J Hum Genet 2019;105:166. Swinnen B, Robberecht W, Van Den Bosch L. RNA toxicity in non-coding repeat expansion disorders. EMBO J 2020;39:e101112. Ueyama M, Nagai Y. Repeat expansion disease models. Adv Exp Med Biol 2018;1076:63. Babi´c Leko M, Župunski V, Kirincich J, et al. Molecular mechanisms of neurodegeneration related to C9orf72 hexanucleotide repeat expansion. Behav Neurol 2019;2019:2909168. Goodman LD, Bonini NM. Repeat-associated non-AUG (RAN) translation mechanisms are running into focus for GGGGCC-repeat associated ALS/FTD. Prog Neurobiol 2019;183:101697. LaCroix AJ, Stabley D, Sahraoui R, et al. GGC repeat expansion and exon 1 methylation of XYLT1 is a common pathogenic variant in Baratela-Scott syndrome. Am J Hum Genet 2019;104:35. Stoyas CA, La Spada AR. The CAG-polyglutamine repeat diseases: a clinical, molecular, genetic, and pathophysiologic nosology. Handb Clin Neurol 2018;147:143. Saade JS, Xing C, Gong X, et al. Instability of TCF4 triplet repeat expansion with parent-child transmission in Fuchs’ endothelial corneal dystrophy. Invest Ophthalmol Vis Sci 2018;59:4065. Nguyen L, Cleary JD, Ranum LPW. Repeat-associated non-ATG translation: molecular mechanisms and contribution to neurological disease. Annu Rev Neurosci 2019;42:227. Semaka A, Kay C, Belfroid RD, et al. A new mutation for Huntington disease following maternal transmission of an intermediate allele. Eur J Med Genet 2015;58:28.

133. Holmans PA, Massey TH, Jones L. Genetic modifiers of Mendelian disease: Huntington’s disease and the trinucleotide repeat disorders. Hum Mol Genet 2017;26:R83. 134. Cooper-Knock J, Shaw PJ, Kirby J. The widening spectrum of C9ORF72-related disease; genotype/phenotype correlations and potential modifiers of clinical phenotype. Acta Neuropathol 2014;127:333. 135. Ida CM, Butz ML, Lundquist PA, Dawson DB. C9orf72 repeat expansion frequency among patients with Huntington disease genetic testing. Neurodegener Dis 2018;18:239. 136. Akimoto C, Volk AE, vanBlitterswijk M, et al. A blinded international study on the reliability of genetic testing for GGGGCC-repeat expansions in C9orf72 reveals marked differences in results among 14 laboratories. J Med Genet 2014;51:419. 137. Rollinson S, Bennion Callister J, Young K, et al. Small deletion in C9orf72 hides a proportion of expansion carriers in FTLD. Neurobiol Aging 2015;36:1601.e1. 138. McFarland KN, Liu J, Landrian I, et al. Repeat interruptions in spinocerebellar ataxia type 10 expansions are strongly associated with epileptic seizures. Neurogenetics 2014;15:59. 139. Hersheson J, Haworth A, Houlden H. The inherited ataxias: genetic heterogeneity, mutation databases, and future directions in research and clinical diagnostics. Hum Mutat 2012;33:1324. 140. Di Gregorio E, Borroni B, Giorgio E, et al. ELOVL5 mutations cause spinocerebellar ataxia 38. Am J Hum Genet 2014;95:209. 141. Seixas AI, Loureiro JR, Costa C, et al. A pentanucleotide ATTTC repeat insertion in the non-coding region of DAB1, mapping to SCA37, causes spinocerebellar ataxia. Am J Hum Genet 2017;101:87. 142. Loureiro JR, Oliveira CL, Mota C, et al. Mutational mechanism for DAB1 (ATTTC)n insertion in SCA37: ATTTT repeat lengthening and nucleotide substitution. Hum Mutat 2019;40:404. 143. Udd B, Krahe R. The myotonic dystrophies: molecular, clinical, and therapeutic challenges. Lancet Neurol 2012;11:891. 144. Wieben ED, Aleff RA, Eckloff BW, et al. Comprehensive assessment of genetic variants within TCF4 in Fuchs’ endothelial corneal dystrophy. Invest Ophthalmol Vis Sci 2014;55:6101. 145. Soliman AZ, Xing C, Radwan SH, et al. Correlation of severity of Fuchs endothelial corneal dystrophy with triplet repeat expansion in TCF4. JAMA Ophthalmol 2015;133:1386.

CHAPTER 14

146. Zhang J, McGhee CNJ, Patel DV. The molecular basis of Fuchs’ endothelial corneal dystrophy. Mol Diagn Ther 2019;23:97. 147. Shoubridge C, Gecz J. Polyalanine tract disorders and neurocognitive phenotypes. Adv Exp Med Biol 2012;769:185. 148. Bishara J, Keens TG, Perez IA. The genetics of congenital central hypoventilation syndrome: clinical implications. Appl Clin Genet. 2018;11:135. 149. Vanneste E, Voet T, Le Caignec C. Chromosome instability is common in human cleavage-stage embryos. Nat Med 2009;15:577. 150. Campbell IM, Shaw CA, Stankiewicz P, et al. Somatic mosaicism: implications for disease and transmission genetics [published correction appears in Trends Genet 2016 Feb;32:138]. Trends Genet 2015;31:382. 151. Bielanska M, Tan SL, Ao A. Chromosomal mosaicism throughout human preimplantation development in vitro: incidence, type, and relevance to embryo outcome. Hum Reprod 2002;17:413. 152. Zhang F, Khajavi M, Connolly AM, et al. The DNA replication FoSTeS/MMBIR mechanism can generate genomic, genic and exonic complex rearrangements in humans. Nat Genet 2009;41:849. 153. Campbell IM, Kolodziejska KE, Quach MM, et al. TGFBR2 deletion in a 20-month-old female with developmental delay and microcephaly. Am J Med Genet A 2011;155A:1442. 154. Frank SA. Somatic mosaicism and disease. Curr Biol 2014;24:R577. 155. Spinner NB, Conlin LK. Mosaicism and clinical genetics. Am J Med Genet C Semin Med Genet 2014;166C:397. 156. Cohen AS, Wilson SL, Trinh J, et al. Detecting somatic mosaicism: considerations and clinical implications. Clin Genet 2015;87:554. 157. Campbell IM, Yuan B, Robberecht C, et al. Parental somatic mosaicism is underrecognized and influences recurrence risk of genomic disorders. Am J Hum Genet 2014;95:173. 158. D’Gama AM, Walsh CA. Somatic mosaicism and neurodevelopmental disease. Nat Neurosci 2018;21:1504. 159. Comstock JM, Putnam AR, Sangle N, et al. Recurrence of achondrogenesis type 2 in sibs: additional evidence for germline mosaicism. Am J Med Genet A 2010;152A:1822. 160. Natacci F, Baffico M, Cavallari U, et al. Germline mosaicism in achondroplasia detected in sperm DNA of the father of three affected sibs. Am J Med Genet A 2008;146A:784.

Molecular Genetics and Prenatal Diagnosis 599

161. Toschi B, Valetto A, Bertini V, et al. Acro-cardio-facial syndrome: a microdeletion syndrome? Am J Med Genet A 2012;158A:1994. 162. Graham GE, MacLeod PM, Lillicrap DP, et al. Gonadal mosaicism in a family with adrenoleukodystrophy: molecular diagnosis of carrier status among daughters of a gonadal mosaic when direct detection of the mutation is not possible. J Inherit Metab Dis 1992;15:68. 163. Wang Y, Busin R, Reeves C, et al. X-linked adrenoleukodystrophy: ABCD1 de novo mutations and mosaicism. Mol Genet Metab 2011;104:160. 164. Aldred MA, Bagshaw RJ, Macdermot K, et al. Germline mosaicism for a GNAS1 mutation and Albright hereditary osteodystrophy. J Med Genet 2000;37:E35. 165. Bachoo S, Gibbons RJ. Germline and gonosomal mosaicism in the ATR-X syndrome. Eur J Hum Genet 1999;7:933. 166. Anazi S, Al-Sabban E, Alkuraya FS. Gonadal mosaicism as a rare cause of autosomal recessive inheritance. Clin Genet 2014;85:278. 167. Beicht S, Strobl-Wildemann G, Rath S, et al. Next generation sequencing as a useful tool in the diagnostics of mosaicism in Alport syndrome. Gene 2013;526:474. 168. Yazaki M, Yamashita T, Kincaid JC, et al. Rapidly progressive amyloid polyneuropathy associated with a novel variant transthyretin serine 25. Muscle Nerve 2002;25:244. 169. Bai Z, Kong X. Extension of the mutation spectrum of PAX6 from three Chinese congenital aniridia families and identification of male gonadal mosaicism. Mol Genet Genomic Med 2018;6:1053. 170. Barbaro V, Nardiello P, Castaldo G, et al. A novel de novo missense mutation in TP63 underlying germline mosaicism in AEC syndrome: implications for recurrence risk and prenatal diagnosis. Am J Genet A 2012;158A:1957. 171. Allanson JE. Germinal mosaicism in Apert syndrome. Clin Genet 1986;29:429. 172. Satoh C, Maekawa R, Kinoshita A, et al. Three brothers with a nonsense mutation in KAT6A caused by parental germline mosaicism. Hum Genome Var 2017;4:17045. 173. Schirwani S, Hauser N, Platt A, et al. Mosaicism in ASXL3-related syndrome: Description of five patients from three families. Eur J Med Genet 2020;63:103925. 174. Bermúdez-López C, García-de Teresa B, González-del Angel A, et al. Germinal mosaicism in a sample of families with Duchenne/Becker muscular dystrophy with partial deletions in the DMD gene. Genet Test Mol Biomarkers 2014;18:93. 175. Robertson SP, Kirk E, Bernier F, et al. Congenital hypertrichosis, osteochondrodysplasia, and

600 Genetic Disorders and the Fetus

176.

177.

178.

179.

180.

181.

182. 183.

184.

185.

186.

187.

cardiomegaly: Cantú syndrome. Am J Med Genet 1999;85:395. Geoghegan S, King G, Henchliffe J, et al. A sibling pair with cardiofaciocutaneous syndrome (CFC) secondary to BRAF mutation with unaffected parents – the first cases of gonadal mosaicism in CFC?. Am J Med Genet A 2018;176:1637. David G, Dürr A, Stevanin G, et al. Molecular and clinical correlations in autosomal dominant cerebellar ataxia with progressive macular dystrophy (SCA7). Hum Mol Genet 1998;7:165. Pauli S, Pieper L, Häberle J, et al. Proven germline mosaicism in a father of two children with CHARGE syndrome. Clin Genet 2009;75:473. Viart V, Willems M, Ishmukhametova A, et al. Germline mosaicism is a pitfall in PGD for X-linked disorders. Single sperm typing detects very low frequency paternal gonadal mosaicism in a case of recurrent chondrodysplasia punctata misattributed to a maternal origin. Prenat Diagn 2017;37:201. Horn D, Delaunoy JP, Kunze J. Prenatal diagnosis in Coffin-Lowry syndrome demonstrates germinal mosaicism confirmed by mutation analysis. Prenat Diagn 2001;21:881. Putnam EA, Park ES, Aalfs CM, et al. Parental somatic and germ-line mosaicism for a FBN2 mutation and analysis of FBN2 transcript levels in dermal fibroblasts. Am J Hum Genet 1997;60:818. Khan AO, Khalil DS, Al Sharif LJ, et al. Fibrosis of the extraocular muscles. Ophthalmology 2010;117:154. Hammel M, Klein M, Trips T, et al. Congenital central hypoventilation syndrome due to PHOX2b gene deficits: inheritance from asymptomatic parents. Klin Padiatr 2009;221:286. Bachetti T, Di Duca M, Della Monica M, et al. Recurrence of CCHS associated PHOX2B poly-alanine expansion mutation due to maternal mosaicism. Pediatr Pulmonol 2014;49:E45. Has C, Bruckner-Tuderman L, Müller D, et al. The Conradi-Hünermann-Happle syndrome (CDPX2) and emopamil binding protein: novel mutations, and somatic and gonadal mosaicism. Hum Mol Genet 2000;9:1951. Niu DM, Huang JY, Li HY, et al. Paternal gonadal mosaicism of NIPBL mutation in a father of siblings with Cornelia de Lange syndrome. Prenat Diagn 2006;26:1054. Sol-Church K, Stabley DL, Demmer LA, et al. Male-to-male transmission of Costello syndrome: G12S HRAS germline mutation inherited from a father with somatic mosaicism. Am J Genet A 2009;149A:315.

188. Goriely A, Lord H, Lim J, et al. Germline and somatic mosaicism for FGFR2 mutation in the mother of a child with Crouzon syndrome: implications for genetic testing in “paternal age-effect” syndromes. Am J Med Genet A 2010;152:2067. 189. Chen XL, Zhao Y, Ke HP, et al. Detection of somatic and germline mosaicism for the LAMP2 gene mutation c.808dupG in a Chinese family with Danon disease. Gene 2012;507:174. 190. Takashima H, Nakagawa M, Kanzaki A, et al. Germline mosaicism of MPZ gene in Dejerine-Sottas syndrome (HMSN III) associated with hereditary stomatocytosis. Neuromuscul Disord 1999;9:232. 191. Melis MA, CauM, Congiu R, et al. Germinal mosaicism in a Duchenne muscular dystrophy family: implications for genetic counselling. Clin Genet 1993;43:247. 192. Xiao H, Zhang Z, Li T, et al. [Germinal Mosaicism for Partial Deletion of the Dystrophin Gene in a Family Affected With Duchenne Muscular Dystrophy]. Zhonghua Yi Xue Yi Chuan Xue Za Zhi 2019;36:1015. 193. Vulliamy TJ, Knight SW, Heiss NS, et al. Dyskeratosis congenita caused by a 3′ deletion: germline and somatic mosaicism in a female carrier. Blood 1999;94:1254. 194. Barrow LL, van Bokhoven H, Daack-Hirsch S, et al. Analysis of the p63 gene in classical EEC syndrome, related syndromes, and non-syndromic orofacial clefts. J Med Genet 2002;39:559. 195. Enriquez A, Krivanek M, Flöttmann R, et al. Recurrence of split hand/foot malformation, cleft lip/palate, and severe urogenital abnormalities due to germline mosaicism for TP63 mutation. Am J Med Genet A. 2016;170:2372. 196. van den Akker PC, Nijenhuis M, Meijer G, et al. Natural gene therapy in dystrophic epidermolysis bullosa. Arch Dermatol 2012;148:213. 197. Pianese L, Fortunato A, Silvestri S, et al. Maternal germline mosaicism in Fabry disease. Neurol Sci 2019;40:1279. 198. Griggs RC, Tawil R, Storvick D, et al. Genetics of facioscapulohumeral muscular dystrophy: new mutations in sporadic cases. Neurology 1993;43:2369. 199. Wieland K, Millar DS, Grundy CB, et al. Molecular genetic analysis of factor X deficiency: gene deletion and germline mosaicism. Hum Genet 1991;86:273. 200. Samadder NJ, Gornick M, Everett J, et al. Inflammatory bowel disease and familial adenomatous polyposis. J Crohns Colitis 2013;7:e103. 201. Macrae F, du Sart D, Nasioulas S. Familial adenomatous polyposis. Best Pract Res Clin Gastroenterol 2009;23:197.

CHAPTER 14

202. Choi HJ, Lee BH, Cho HY, et al. Familial focal segmental glomerulosclerosis associated with an ACTN4 mutation and paternal germline mosaicism. Am J Kidney Dis 2008;51:834. 203. Forissier JF, Richard P, Briault S, et al. First description of germline mosaicism in familial hypertrophic cardiomyopathy. J Med Genet 2000;37:132. 204. Janoff HB, Muenke M, Johnson LO, et al. Fibrodysplasia ossificans progressiva in two half-sisters: evidence for maternal mosaicism. Am J Med Genet 1996;61:320. 205. Prior TW, Papp AC, Snyder PJ, et al. Germline mosaicism at the fragile X locus. Am J Med Genet 1995;55:384. 206. Chak WL, To KF, Cheng YL, et al. Gonadal mosaicism of Frasier syndrome in 3 Chinese siblings with donor splice site mutation of Wilms’ tumour gene. Nephron 2002;91:526. 207. Bengani H, Handley M, Alvi M, et al. Clinical and molecular consequences of disease-associated de novo mutations in SATB2. Genet Med 2017;19:900. 208. Stoppa-Vaucher S, Ayabe T, Paquette J, et al. 46, XY gonadal dysgenesis: new SRY point mutation in two siblings with paternal germ line mosaicism. Clin Genet 2012;82:505. 209. Courage C, Jackson CB, Owczarek-Lipska M, et al. Novel synonymous and missense variants in FGFR1 causing Hartsfield syndrome. Am J Med Genet A 2019;179:2447. 210. Sommer SS, Scaringe WA, Hill KA. Human germline mutation in the factor IX gene. Mutat Res 2001;487:1. 211. Cutler JA, Mitchell MJ, Smith MP, et al. Germline mosaicism resulting in the transmission of severe hemophilia B from a grandfather with a mild deficiency. Am J Med Genet A 2004;129A:13. 212. Ponard D, Gaboriaud C, Charignon D, et al. SERPING1 mutation update: mutation spectrum and C1 Inhibitor phenotypes. Hum Mutat 2020;41:38. 213. Eyries M, Coulet F, Girerd B, et al. ACVRL1 germinal mosaic with two mutant alleles in hereditary hemorrhagic telangiectasia associated with pulmonary arterial hypertension. Clin Genet 2012;82:173. 214. Cserhalmi-Friedman PB, Anyane-Yeboa K, Christiano AM. Paternal germline mosaicism in Herlitz junctional epidermolysis bullosa. Exp Dermatol 2002;11:468. 215. Braulke I, Herzog S, Thies U, et al. Holt-Oram syndrome in four half-siblings with unaffected parents: brief clinical report. Clin Genet 1991;39:241. 216. Ben Simon-Schiff E, Bach G, Zlotogora J, et al. Combined enzymatic and linkage analysis for heterozygote detection in Hunter syndrome: identification of an apparent case of germinal mosaicism. Am J Med Genet 1993;47:837.

Molecular Genetics and Prenatal Diagnosis 601

217. Lin Y, Cai Y, Xu J, et al. ‘Isolated’ germline mosaicism in the phenotypically normal father of a girl with X-linked hypophosphatemic rickets. Eur J Endocrinol 2020;182:K1. 218. Kirchman TT, Levy ML, Lewis RA, et al. Gonadal mosaicism for incontinentia pigmenti in a healthy male. J Med Genet 1995;32:887. 219. Wong SC, Cobben JM, Hiemstra S, et al. Karsch-Neugebauer syndrome in two sibs with unaffected parents. Am J Med Genet 1998;75:207. 220. Sbidian E, Feldmann D, Bengoa J, et al. Germline mosaicism in keratitis-ichthyosis-deafness syndrome: pre-natal diagnosis in a familial lethal form. Clin Genet 2010;77:587. 221. Vos YJ, de Walle HE, Bos KK, et al. Genotype–phenotype correlations in L1 syndrome: a guide for genetic counselling and mutation analysis. J Med Genet 2010;47:169. 222. Matsumoto N, Leventer RJ, Kuc JA, et al. Mutation analysis of the DCX gene and genotype/phenotype correlation in subcortical band heterotopia. Eur J Hum Genet 2001;9:5. 223. Perez Maturo J, Vega P, Medina N, et al. Inferring parental gonadal mosaicism in LMNA-associated muscular dystrophy by ultra-deep next generation sequencing: A sensitive approach providing valuable information for genetic counseling. Am J Med Genet A 2019;179:1074. 224. Hancarova M, Havlovicova M, Putzova M, et al. Parental gonadal but not somatic mosaicism leading to de novo NFIX variants shared by two brothers with Malan syndrome. Am J Med Genet A 2019;179:2119. 225. Rivière JB, Mirzaa GM, O’Roak BJ, et al. De novo germline and postzygotic mutations in AKT3, PIK3R2, and PIK3CA cause a spectrum of related megalencephaly syndromes. Nat Genet 2012;44:934. 226. Di Donato N, Klink B, Hahn G, et al. Interstitial deletion 1p36.32 in two brothers with a distinct phenotype – overgrowth, macrocephaly and nearly normal intellectual function. Eur J Med Genet 2014;57:494. 227. Tang HS, Wang DG, Xie XM, et al. Apparent germline mosaicism for a 15q11-q13 deletion causing recurrent Angelman syndrome in a Chinese family. Eur J Obstet Gynecol Reprod Biol 2019;236:255. 228. Nimmakayalu M, Horton VK, Darbro B, et al. Apparent germline mosaicism for a novel 19p13.13 deletion disrupting NFIX and CACNA1A. Am J Med Genet A 2013;161A:1105. 229. Vincent MC, Guiraud-Chaumeil C, Laporte J, et al. Extensive germinal mosaicismin a family with X linked myotubular myopathy simulates genetic heterogeneity. J Med Genet 1998;35:241.

602 Genetic Disorders and the Fetus

230. Häne BG, Rogers RC, Schwartz CE. Germline mosaicism in X-linked myotubular myopathy. Clin Genet 1999;56:77. 231. Seidahmed MZ, Salih MA, Abdelbasit OB, et al. Gonadal mosaicism for ACTA1 gene masquerading as autosomal recessive nemaline myopathy. Am J Med Genet A 2016;170:2219. 232. Haack TB, Hogarth P, Kruer MC, et al. Exome sequencing reveals de novo WDR45 mutations causing a phenotypically distinct, X-linked dominant form of NBIA. Am J Hum Genet 2012;91:1144. 233. Menke LA, Engelen M, Alders M, et al. Recurrent GNAO1 mutations associated with developmental delay and a movement disorder. J Child Neurol 2016;31:1598. 234. Cross LA, McWalter K, Keller-Ramey J, et al. A report of gonadal mosaicism in DHX30-related neurodevelopmental disorder. Clin Dysmorphol 2020;29:161. 235. Bottillo I, Torrente I, Lanari V, et al. Germline mosaicism in neurofibromatosis type 1 due to a paternally derived multi-exon deletion. Am J Med Genet A 2010;152A:1467. 236. Muram-Zborovski TM, Vaughn CP, Viskochil DH, et al. NF1 exon 22 analysis of individuals with the clinical diagnosis of neurofibromatosis type 1. Am J Med Genet A 2010;152A:1973. 237. Trevisson E, Forzan M, Salviati L, et al. Neurofibromatosis type 1 in two siblings due to maternal germline mosaicism. Clin Genet 2014;85:386. 238. Sestini R, Vivarelli R, Balestri P, et al. Neurofibromatosis type 2 attributable to gonosomal mosaicism in a clinically normal mother, and identification of seven novel mutations in theNF2 gene. Hum Genet 2000;107:366. 239. Aoki Y, Niihori T, Banjo T, et al. Gain-of-function mutations in RIT1 cause Noonan syndrome, a RAS/MAPK pathway syndrome. Am J Hum Genet 2013;93:173. 240. Hafner C, Groesser L. Mosaic RASopathies. Cell Cycle 2013;12:43. 241. Satre V, Monnier N, Berthoin F, et al. Characterization of a germline mosaicism in families with Lowe syndrome, and identification of seven novel mutations in the OCRL1 gene. Am J Hum Genet 1999;65:68. 242. Redwood A, Douzgou S, Waller S, et al. Congenital cataracts in females caused by BCOR mutations; report of six further families demonstrating clinical variability and diverse genetic mechanisms. Eur J Med Genet 2020;63:103658. 243. Bowling F, McGown I, McGill J, et al. Maternal gonadal mosaicism causing ornithine transcarbamylase deficiency. Am J Med Genet 1999;85:452.

244. Chen CP, Lin SP, Su YN, et al. Prenatal diagnosis of recurrent autosomal dominant osteogenesis imperfecta associated with unaffected parents and paternal gonadal mosaicism. Taiwan J Obstet Gynecol 2013;52:106. 245. O’Byrne JJ, Phelan E, Steenackers E, et al. Germline mosaicism in osteopathia striata with cranial sclerosis – recurrence in siblings. Clin Dysmorphol 2016;25:45. 246. Robertson SP, Thompson S, Morgan T, et al. Postzygotic mutation and germline mosaicism in the otopalatodigital syndrome spectrum disorders. Eur J Hum Genet 2006;14:549. 247. Ng D, Johnston JJ, Turner JT, et al. Gonadal mosaicism in severe Pallister-Hall syndrome. Am J Med Genet A 2004;124A:296. Erratum in: Am J Med Genet A 2005;136:225. 248. Iliuta IA, Kalatharan V, Wang K, et al. Polycystic kidney disease without an apparent family history. J Am Soc Nephrol 2017;28:2768. 249. Buffet A, Smati S, Mansuy L, et al. Mosaicism in HIF2A related polycythemia-paraganglioma syndrome. J Clin Endocrinol Metab 2014;99:e369. 250. Hall JG, Dorst JP, Rotta J, et al. Gonadal mosaicism in pseudoachondroplasia. Am J Med Genet 1987;28:143. 251. Amiel J, Audollent S, Joly D, et al. PAX2 mutations in renal-coloboma syndrome: mutational hotspot and germline mosaicism. Eur J Hum Genet 2000;8:820. 252. Barbosa RH, Vargas FR, Aguiar FC, et al. Hereditary retinoblastoma transmitted by maternal germline mosaicism. Pediatr Blood Cancer 2008;51:598. 253. Astudillo PP, Chan HS, Héon E, et al. Late-diagnosis retinoblastoma with germline mosaicism in an 8-yearold. J AAPOS 2014;18:500. 254. Armstrong J, Aibar E, Pineda M, et al. Prenatal diagnosis in Rett syndrome. Fetal Diagn Ther 2002;17:200. 255. Evans JC, Archer HL, Whatley SD, et al. Germline mosaicism for a MECP2 mutation in a man with two Rett daughters. Clin Genet 2006;70:336. 256. McMahon KQ, Papandreou A, Ma M, et al. Familial recurrences of FOXG1-related disorder: evidence for mosaicism. Am J Med Genet A. 2015;167A:3096. 257. Bourdeaut F, Lequin D, Brugières L, et al. Frequent hSNF5/INI1 germline mutations in patients with rhabdoid tumor. Clin Cancer Res 2011;17:31. 258. Eaton KW, Tooke LS, Wainwright LM, et al. Spectrum of SMARCB1/INI1 mutations in familial and sporadic rhabdoid tumors. Pediatr Blood Cancer 2011;56:7.

CHAPTER 14

259. Gauthier J, Champagne N, Lafrenière RG, et al. De novo mutations in the gene encoding the synaptic scaffolding protein SHANK3 in patients ascertained for schizophrenia. Proc Natl Acad Sci U S A 2010;107:7863. 260. O’Marcaigh AS, Puck JM, Pepper AE, et al. Maternal mosaicism for a novel interleukin-2 receptor gamma-chain mutation causing X-linked severe combined immunodeficiency in a Navajo kindred. J Clin Immunol 1997;17:29. 261. Shanske AL, Goodrich JT, Ala-Kokko L, et al. Germline mosaicism in Shprintzen-Goldberg syndrome. Am J Med Genet A 2012;158A:1574. 262. Gordo G, Tenorio J, Arias P, et al. mTOR mutations in Smith-Kingsmore syndrome: Four additional patients and a review. Clin Genet 2018;93:762. 263. de Ravel TJ, De Smet L, Fryns JP. Apparently new autosomal dominant spondyloepimetaphyseal dysplasia: gonadal mosaicism onset. Clin Dysmorphol 2002;11:261. 264. Kamien B, Dadd T, Buckman M, et al. Somatic-gonadal mosaicism causing Sotos syndrome. Am J Med Genet A 2016;170:3360. 265. Yates JR, van Bakel I, Sepp T, et al. Female germline mosaicism in tuberous sclerosis confirmed by molecular genetic analysis. Hum Mol Genet 1997;6:2265. 266. Rose VM, Au KS, Pollom G, et al. Germ-line mosaicism in tuberous sclerosis: how common? Am J Hum Genet 1999;64:986. 267. Murray EW, Giles AR, Lillicrap D. Germ-line mosaicism for a valine-to-methionine substitution at residue 553 in the glycoprotein Ib-binding domain of von Willebrand factor, causing type IIB von Willebrand disease. Am J Hum Genet 1992;50:199. 268. Kapur S, Karam S. Germ-line mosaicism in Waardenburg syndrome. Clin Genet 1991;39:194. 269. Bocángel MAP, Melo US, Alves LU, et al. Waardenburg syndrome: Novel mutations in a large Brazilian sample. Eur J Med Genet 2018;61:348. 270. Arveiler B, de Saint-Basile G, Fischer A, et al. Germline mosaicism simulates genetic heterogeneity in Wiskott-Aldrich syndrome. Am J Hum Genet 1990;46:906. 271. Campbell IM, Stewart JR, James RA, et al. Parent of origin, mosaicism, and recurrence risk: probabilistic modeling explains the broken symmetry of transmission genetics. Am J Hum Genet 2014;95:345. 272. Goldmann JM, Wong WS, Pinelli M, et al. Parent-of-origin-specific signatures of de novo mutations [published correction appears in Nat Genet 2018 Nov;50:1615]. Nat Genet 2016;48:935.

Molecular Genetics and Prenatal Diagnosis 603

273. Acuna-Hidalgo R, Veltman JA, Hoischen A. New insights into the generation and role of de novo mutations in health and disease. Genome Biol 2016;17:241. 274. Dong S, Walker MF, Carriero NJ, et al. De novo insertions and deletions of predominantly paternal origin are associated with autism spectrum disorder. Cell Rep 2014;9:16. 275. Erickson RP. Somatic gene mutation and human disease other than cancer: an update. Mutat Res 2010;705:96. 276. Kehrer-Sawatzki H, Kluwe L, Sandig C, et al. High frequency of mosaicism among patients with neurofibromatosis type 1 (NF1) with microdeletions caused by somatic recombination of the JJAZ1 gene. Am J Hum Genet 2004;75:410. 277. Moyhuddin A, Baser ME, Watson C, et al. Somatic mosaicism in neurofibromatosis 2: prevalence and risk of disease transmission to offspring. J Med Genet 2003;40:459. 278. Kluwe L, Mautner V, Heinrich B, et al. Molecular study of frequency of mosaicism in neurofibromatosis 2 patients with bilateral vestibular schwannomas. J Med Genet 2003;40:109. 279. Pham J, Shaw C, Pursley A, et al. Somatic mosaicism detected by exon-targeted, high-resolution aCGH in 10,362 consecutive cases. Eur J Hum Genet 2014;22:969. 280. Kline AD, Moss JF, Selicorni A, et al. Diagnosis and management of Cornelia de Lange syndrome: first international consensus statement. Nat Rev Genet 2018;19:649. 281. Dowsett L, Porras AR, Kruszka P, et al. Cornelia de Lange syndrome in diverse populations. Am J Med Genet A. 2019;179:150. 282. Huisman SA, Redeker EJ, Maas SM, et al. High rate of mosaicism in individuals with Cornelia de Lange syndrome. J Med Genet 2013;50:339. 283. Braunholz D, Obieglo C, Parenti I, et al. Hidden mutations in Cornelia de Lange syndrome limitations of Sanger sequencing in molecular diagnostics. Hum Mutat 2015;36:26. 284. Ansari M, Poke G, Ferry Q, et al. Genetic heterogeneity in Cornelia de Lange syndrome (CdLS) and CdLS-like phenotypes with observed and predicted levels of mosaicism. J Med Genet 2014;5:659. 285. Gripp KW, Stabley DL, Nicholson L, et al. Somatic mosaicism for an HRAS mutation causes Costello Syndrome. Am J Med Genet A 2006;140:2163. 286. Miyatake S, Koshimizu E, Hayashi YK, et al. Deep sequencing detects very-low-grade somatic mosaicism in the unaffected mother of siblings with nemaline myopathy. Neuromuscul Disord 2014;24:642.

604 Genetic Disorders and the Fetus

287. Sigaudy S, Fredouille C, Gambarelli D, et al. Prenatal ultrasonic findings in Proteus syndrome. Prenat Diagn 1998;18:1091. 288. Tissot H, Maugey B, Serville F, et al. Diagnostic antenatal d’un lymphangiome kystique abdomino-pelvien s’intégrant dans le cadre d’un syndrome proteus. [Prenatal diagnosis of abdomino-pelvic cystic lymphangioma as part of proteus syndrome.] J Gynecol Obstet Biol Reprod (Paris) 1991;20:335. 289. Goss JA, Konczyk DJ, Smits P, et al. Diffuse capillary malformation with overgrowth contains somatic PIK3CA variants. Clin Genet 2020; 97:736. 290. Dobyns WB, Mirzaa GM. Megalencephaly syndromes associated with mutations of core components of the PI3K-AKT-MTOR pathway: PIK3CA, PIK3R2, AKT3, and MTOR. Am J Med Genet C Semin Med Genet 2019;181:582. 291. Castillo SD, Baselga E, Graupera M. PIK3CA mutations in vascular malformations. Curr Opin Hematol 2019;26:170. 292. Martinez-Lopez A, Blasco-Morente G, Perez-Lopez I, et al. CLOVES syndrome: review of a PIK3CA-related overgrowth spectrum (PROS). Clin Genet 2017;91:14. 293. Keppler-Noreuil KM, Rios JJ, Parker VE, et al. PIK3CA-related overgrowth spectrum (PROS): diagnostic and testing eligibility criteria, differential diagnosis, and evaluation. Am J Med Genet A 2015;167A:287. 294. Jonkman MF, Pasmooij AMG. Revertant mosaicism – patchwork in the skin. N Eng J Med 2009;360:1680. 295. Inbar-Feigenberg M, Choufani S, Butcher DT, et al. Basic concepts of epigenetics. Fertil Steril 2013;99:607. 296. Bhoj EJ, Rajabi F, Baker SW, et al. Imprinted genes in clinical exome sequencing: Review of 538 cases and exploration of mouse-human conservation in the identification of novel human disease loci. Eur J Med Genet 2020;63:103903. 297. Weksberg R. Imprinted genes and human disease. Am J Med Genet C Semin Med Genet 2010;154:317. 298. Horsthemke B. Mechanisms of imprint dysregulation. Am J Med Genet C Semin Med Genet 2010;154:321. 299. Choufani S, Shapiro JS, Susiarjo M, et al. A novel approach identifies new differentially methylated regions (DMRs) associated with imprinted genes. Genome Res 2011;21:465. 300. Hales BF, Grenier L, Lalancette C, et al. Epigenetic programming: from gametes to blastocyst. Birth Defects Res A Clin Mol Teratol 2011;91:652. 301. Ishida M, Moore GE. The role of imprinted genes in humans. Mol Aspects Med 2013;34:826.

302. Docherty LD, Rezwan FI, Poole RL, et al. Genome-wide DNA methylation analysis of patients with imprinting disorders identifies differentially methylated regions associated with novel candidate imprinted genes. J Med Genet 2014;51:229. 303. Clayton-Smith J, Laan L. Angelman syndrome: a review of the clinical and genetic aspects. J Med Genet 2003;40:87. 304. Dindot SV, Antaiffy BA, Bhattacharjee MB, et al. The Angelman syndrome ubiquitin ligase localizes to the synapse and nucleus and maternal deficiency results in abnormal dendritic spinemorphology. Hum Mol Genet 2008;17:111. 305. Walter J, Paulsen M. Imprinting and disease. Semin Cell Dev Biol 2003;14:101. 306. Rossignol S, Steunou V, Chalas C, et al. The epigenetic imprinting defect of patients with Beckwith–Wiedemann syndrome born after assisted reproductive technology is not restricted to the 11p15 region. J Med Genet 2006;43:902. 307. Wilkins-Haug L, Porter A, Hawley P, et al. Isolated fetal omphalocele, Beckwith–Wiedemann syndrome and assisted reproductive technologies. Birth Defects Res A Clin Mol Teratol 2009;85:58. 308. Barel O, Shalev SA, Ofir R, et al. Maternally inherited Birk Barel mental retardation dysmorphism syndrome caused by a mutation in the genomically imprinted potassium channel KCNK9. Am J Hum Genet 2008;83:193. 309. Fournet JC, Mayaud C, de Lonlay P, et al. Loss of imprinted genes and paternal SUR1 mutations lead to focal form of congenital hyperinsulinism. Horm Res 2000;53:2. 310. Passos-Bueno MR, Cerqueira A, Vainzof M, et al. Myotonic dystrophy: genetic, clinical and molecular analysis of patients from 41 Brazilian families. J Med Genet 1995;32:14. 311. Judson H, Hayward BE, Sheridan E, et al. A global disorder of imprinting in the human female germ line. Nature 2002;416:539. 312. Müller U. Pathological mechanisms and parent-of-origin effects in hereditary paraganglioma/pheochromocytoma (PGL/PCC). Neurogenetics. 2011;12:175. 313. Muller B, Hedrich K, Kock N, et al. Evidence that paternal expression of the epsilon-sarcoglycan gene accounts for reduced penetrance in myoclonus-dystonia. Am J Hum Genet 2002;71:1303. 314. Davies W, Isles AR, Wilkinson LS. Imprinted genes and mental dysfunction. Ann Med 2001;33:428. 315. Fokstuen S, Ginsburg C, Zachmann M, et al. Maternal uniparental disomy 14 as a cause of intrauterine

CHAPTER 14

316.

317.

318.

319.

320.

321.

322.

323.

324.

325.

326.

327.

328.

growth retardation and early onset of puberty. J Pediatr 1999;134:689. Eggermann T, Zerres K, Eggermann K, et al. Uniparental disomy: clinical indications for testing in growth retardation. Eur J Pediatr 2002;161: 305. Azzi S, Brioude F, Le Bouc Y, et al. Human imprinting anomalies in fetal and childhood growth disorders: clinical implications and molecular mechanisms. Curr Pharm Des 2014;20:1751. Temple IK, Shrubb V, Lever M, et al. Isolated imprinting mutation of the DLK1/GTL2 locus associated with a clinical presentation of maternal uniparental disomy of chromosome 14. BMJ Case Rep 2009;06:1997. Irving MD, Buiting K, Kanber D, et al. Segmental paternal uniparental disomy (patUPD) of 14q32 with abnormal methylation elicits the characteristic features of complete patUPD14. Am J Med Genet A 2010;152:1942. Perk J, Makedonski K, Lande L, et al. The imprinting mechanism of the Prader–Willi/Angelman regional control center. EMBO J 2002;21:5807. Shore EM, Ahn J, Jan de Beur S, et al. Paternally inherited inactivating mutations of the GNAS1 gene in progressive osseous heteroplasia. N Engl J Med 2002;346:99. Bastepe M, Fröhlich LF, Hendy GN, et al. Autosomal dominant pseudohypoparathyroidism type Ib is associated with a heterozygous microdeletion that likely disrupts a putative imprinting control element of GNAS. J Clin Invest 2003;112:1255. Bastepe M, Juppner H. Pseudohypoparathyroidism: new insights into an old disease. Endocrinol Metab Clin North Am 2000;29:569. Hannula K, Lipsanen-NymanM,Kristo P, et al. Genetic screening for maternal uniparental disomy of chromosome 7 in prenatal and postnatal growth retardation of unknown cause. Pediatrics 2002;109:441. Netchine I, Rossignol S, Dufourg MN, et al. 11p15 imprinting center region 1 loss of methylation is a common and specific cause of typical Russell–Silver syndrome: clinical scoring system and epigenetic– phenotypic correlations. J Clin Endocrinol Metab 2007;92:3148. Fountain MD, Schaaf CP. Prader-Willi syndrome and Schaaf-Yang syndrome: neurodevelopmental diseases intersecting at the MAGEL2 gene. Diseases 2016;4:2. Amor DJ, Halliday J. A review of known imprinting syndromes and their association with assisted reproduction technologies. Hum Reprod 2008;23:2826. Temple IK, Shield JP. 6q24 transient neonatal diabetes. Rev Endocr Metab Disord 2010;11:199.

Molecular Genetics and Prenatal Diagnosis 605

329. Sensi A, Cavani S, Villa N, et al. Nonhomologous Robertsonian translocations (NHTRs) and uniparental disomy (UPD) risk: an Italian multicentric prenatal survey. Prenat Diagn 2004;24:647. 330. Moradkhani K, Cuisset L, Boisseau P, et al. Risk estimation of uniparental disomy of chromosome 14 or 15 in a fetus with a parent carrying a non-homologous Robertsonian translocation. Should we still perform prenatal diagnosis? Prenat Diagn 2019;39:986. 331. Eggermann T, Soellner L, Buiting K, et al. Mosaicism and uniparental disomy in prenatal diagnosis. Trends Mol Med 2015;21:77. 332. Cortessis VK, Azadian M, Buxbaum J, et al. Comprehensive meta-analysis reveals association between multiple imprinting disorders and conception by assisted reproductive technology. J Assist Reprod Genet 2018;35:943. 333. Savage T, Peek J,Hofman PL, et al. Childhood outcomes of assisted reproductive technology. Hum Reprod 2011;26:2392. 334. Hiura H, Okae H, Miyauchi N, et al. Characterization of DNA methylation errors in patients with imprinting disorders conceived by assisted reproduction technologies. Hum Reprod 2012;27:2541. 335. Vermeiden JPW, Bernardus RE. Are imprinting disorders more prevalent after human in vitro fertilization or intracytoplasmic sperm injection? Fertil Steril 2013;99:642. 336. Eroglu A, Layman LC. Role of ART in imprinting disorder. Semin Reprod Med 2012;30:10.1055/s-0032. 337. Li J, Jia X, Li S, et al. Mutation survey of candidate genes in 40 Chinese patients with congenital ectopia lentis. Mol Vis 2014;20:1017. 338. Willis, BR, Lee, M, Rethanavelu, K, et al. A case of G1013R FBN1 mutation: a potential genotype–phenotype correlation in severe Marfan syndrome. Am J Med Genet Part A 2020;182A:1329. 339. Pepin MG, Schwarze U, Rice KM, et al. Survival is affected by mutation type and molecular mechanism in vascular Ehlers-Danlos syndrome (EDS type IV). Genet Med 2014;16:881. 340. Wang Z, Milunsky J, Yamin M, et al. Analysis by mass spectrometry of 100 cystic fibrosis gene mutations in 92 patients with congenital bilateral absence of the vas deferens. Hum Reprod 2002;17:2066. 341. Thauvin-Robinet C, Munck A, Huet F, et al. CFTR p.Arg117His associated with CBAVD and other CFTR related disorders. J Med Genet 2013;50:220. 342. Xu X, Zheng J, Liao Q, et al. Meta-analyses of 4 CFTR variants associated with the risk of the congenital bilateral absence of the vas deferens. J Clin Bioinforma 2014;4:11.

606 Genetic Disorders and the Fetus

343. de Tayrac M, Roth MP, Jouanolle AM, et al. Genomewide association study identifies TF as a significant modifier gene of iron metabolism in HFE hemochromatosis. J Hepatol 2015;62:664. 344. Gallati S. Disease-modifying genes and monogenic disorders: experience in cystic fibrosis. Appl Clin Genet 2014;7:133. 345. Bandiera S, Hatem E, Lyonnet S, et al. MicroRNAs in diseases: from candidate to modifier genes. Clin Genet 2010;77:306. 346. Costanzo M, Kuzmin E, van Leeuwen J, et al. Global genetic networks and the genotype-to-phenotype relationship. Cell 2019;177:85. 347. Deltas C. Digenic inheritance and genetic modifiers. Clin Genet 2018;93:429. 348. Yang Y, Muzny DM, Xia F, et al. Molecular findings among patients referred for clinical whole-exome sequencing. JAMA 2014;312:1870. 349. Lupski JR, Belmont JW, Boerwinkle E, et al. Clan genomics and the complex architecture of human disease. Cell 2011;147:32. 350. Ochoa JP, Lopes LR, Perez-Barbeito M, et al. Deletions of specific exons of FHOD3 detected by next-generation sequencing are associated with hypertrophic cardiomyopathy. Clin Genet 2020;98:86. 351. Ingles J, Sarina T, Yeates L, et al. Clinical predictors of genetic testing outcomes in hypertrophic cardiomyopathy. Genet Med 2013;15:972. 352. Sattar Y, Abdullah HM, Neisani Samani E, et al. Arrhythmogenic right ventricular cardiomyopathy/dysplasia: an updated review of diagnosis and management. Cureus 2019;11:e5381. 353. Marcus FI, Edson S, Towbin JA. Genetics of arrhythmogenic right ventricular cardiomyopathy: a practical guide for physicians. J Am Coll Cardiol 2013;61:1945. 354. Stevenson VA, Ito M, Milunsky JM. Connexin-30 deletion analysis in connexin-26 heterozygotes. Genet Test 2003;7:151. 355. Pandya A, O’Brien A, Kovasala M, et al. Analyses of del(GJB6-D13S1830) and del(GJB6-D13S1834) deletions in a large cohort with hearing loss: caveats to interpretation of molecular test results in multiplex families. Mol Genet Genomic Med 2020;8:e1171. 356. Schrauwen I, Chakchouk I, Acharya A, et al. Novel digenic inheritance of PCDH15 and USH1G underlies profound non-syndromic hearing impairment. BMC Med Genet 2018;19:122. 357. Manara E, Paolacci S, D’Esposito F, et al. Mutation profile of BBS genes in patients with Bardet-Biedl syndrome: an Italian study. Ital J Pediatr 2019;45:72.

358. van Rij MC, de Die-Smulders CE, Bijlsma EK, et al. Evaluation of exclusion prenatal and exclusion preimplantation genetic diagnosis for Huntington’s disease in the Netherlands. Clin Genet 2013;83:118. 359. He X, Sanders SJ, Liu L, et al. Integrated model of de novo and inherited genetic variants yields greater power to identify risk genes. PLoS Genet 2013;9:e1003671. 360. Georgieva L, Rees E, Moran JL, et al. De novo CNVs in bipolar affective disorder and schizophrenia. Hum Mol Genet 2014;23:6677. 361. Kloiber S, Rosenblat JD, Husain MI, et al. Neurodevelopmental pathways in bipolar disorder. Neurosci Biobehav Rev 2020;112:213. 362. Turner TN, Hormozdiari F, Duyzend MH, et al. Genome sequencing of autism-affected families reveals disruption of putative noncoding regulatory DNA. Am J Hum Genet 2016;98:58. 363. Fernandez A, Drozd MM, Thümmler S, et al. Childhood-onset schizophrenia: a systematic overview of its genetic heterogeneity from classical studies to the genomic era. Front Genet 2019;10:1137. 364. Savova V, Vinogradova S, Pruss D, et al. Risk alleles of genes with monoallelic expression are enriched in gain-of-function variants and depleted in loss-of-function variants for neurodevelopmental disorders. Mol Psychiatry 2017;22:1785. 365. Sanders SJ, Murtha MT, Gupta AR, et al. De novo mutations revealed by whole-exome sequencing are strongly associated with autism. Nature 2012;485:237. 366. O’Roak BJ, Vives L, Girirajen S, et al. Sporadic autism exomes reveal a highly interconnected protein network of de novo mutations. Nature 2012;485:246. 367. Iossifov I, O’Roak BJ, Sanders SJ, et al. The contribution of de novo coding mutations to autism spectrum disorder. Nature 2014;515:216. 368. Codina-Solà M, Rodríguez-Santiago B, Homs A, et al. Integrated analysis of whole-exome sequencing and transcriptome profiling in males with autism spectrum disorders. Mol Autism 2015;6:21. 369. Callaghan DB, Rogic S, Tan PPC, et al. Whole genome sequencing and variant discovery in the ASPIRE autism spectrum disorder cohort. Clin Genet 2019;96:199. 370. Leblond CS, Heinrich J, Delorme R, et al. Genetic and functional analyses of SHANK2 mutations suggest a multiple hit model of autism spectrum disorders. PLoS Genet 2012;8:e1002521. 371. Bacchelli E, Cameli C, Viggiano M, et al. An integrated analysis of rare CNV and exome variation in autism spectrum disorder using the Infinium PsychArray. Sci Rep 2020;10:3198.

CHAPTER 14

372. Ashitha SNM, Ramachandra NB. Integrated functional analysis implicates syndromic and rare copy number variation genes as prominent molecular players in pathogenesis of autism spectrum disorders. Neuroscience 2020;438:25. 373. Klopocki E, Schulze H, Strauss G, et al. Complex inheritance pattern resembling autosomal recessive inheritance involving a microdeletion in thrombocytopenia-absent radius syndrome. Am J Hum Genet 2007;80:232. 374. Boussion S, Escande F, Jourdain AS, et al. TAR syndrome: clinical and molecular characterization of a cohort of 26 patients and description of novel noncoding variants of RBM8A. Hum Mutat 2020;41:1220. 375. Albers CA, Paul DS, Schulze H, et al. Compound inheritance of a low-frequency regulatory SNP and a rare null mutation in exon-junction complex subunit RBM8A causes TAR syndrome. Nat Genet 2012;44:435. 376. Tongsong T, Sirichotiyakul S, Chanprapaph P. Prenatal diagnosis of thrombocytopenia-absent-radius (TAR) syndrome. Ultrasound Obstet Gynecol. 2000;15:256. 377. Travessa AM, Dias P, Santos A, et al. Upper limb phocomelia: A prenatal case of thrombocytopenia-absent radius (TAR) syndrome illustrating the importance of chromosomal microarray in limb reduction defects. Taiwan J Obstet Gynecol 2020;59:318. 378. Papoulidis I, Oikonomidou E, Orru S, et al. Prenatal detection of TAR syndrome in a fetus with compound inheritance of an RBM8A SNP and a 334-kb deletion: a case report. Mol Med Rep 2014;9:163. 379. Tsai HF, Liu CS, Chen GD, et al. Prenatal diagnosis of Machado-Joseph disease/spinocerebellar ataxia type 3 in Taiwan: early detection of expanded ataxin-3. J Clin Lab Anal 2003;17:195. 380. Cruz-Mariño T, Velázquez-Pérez L, Gonzéles-Zaldivar Y, et al. The Cuban program for predictive testing of SCA2: 11 years and 768 individuals to learn from. Clin Genet 2013;83:518. 381. Cruz-Mariño T, Laffita-Mesa JM, Gonzáles-Zaldivar Y, et al. Large normal and intermediate alleles in the context of SCA2 prenatal diagnosis. J Genet Couns 2014;23:89. 382. Cahn S, Rosen A, Wilmot G. Spinocerebellar ataxia patient perceptions regarding reproductive options. Mov Disord Clin Pract 2019;7:37. 383. Ishige T, Sawai S, Itoga S, et al. Pentanucleotide repeat-primed PCR for genetic diagnosis of spinocerebellar ataxia type 31. J Hum Genet 2012;57:807. 384. Cagnoli C, Stevanin G, Michielotto C, et al. Large pathogenic expansions in the SCA2 and SCA7 genes

385.

386.

387.

388.

389.

390.

391.

392.

393.

394.

395.

396.

397.

398.

399.

Molecular Genetics and Prenatal Diagnosis 607

can be detected by fluorescent repeat-primed polymerase chain reaction assay. J Mol Diagn 2006;8:128. Carvill GL, Helbig KL, Myers CT, et al. Damaging de novo missense variants in EEF1A2 lead to a developmental and degenerative epileptic-dyskinetic encephalopathy. Hum Mutat 2020;41:1263. Epi25 Collaborative. Ultra-rare genetic variation in the epilepsies: a whole-exome sequencing study of 17,606 individuals. Am J Hum Genet 2019;105:267. Dunn P, Albury CL, Maksemous N, et al. Next generation sequencing methods for diagnosis of epilepsy syndromes. Front Genet 2018;9:20. Tumien˙e B, Maver A, Writzl K, et al. Diagnostic exome sequencing of syndromic epilepsy patients in clinical practice. Clin Genet 2018;93:1057. Helbig KL, Farwell Hagman KD, Shinde DN, et al. Diagnostic exome sequencing provides a molecular diagnosis for a significant proportion of patients with epilepsy. Genet Med 2016;18:898. Demos M, Guella I, DeGuzman C, et al. Diagnostic yield and treatment impact of targeted exome sequencing in early-onset epilepsy. Front Neurol 2019;10:434. Tsang MH, Leung GK, Ho AC, et al. Exome sequencing identifies molecular diagnosis in children with drug-resistant epilepsy. Epilepsia Open 2018;4:63. Li J, Gao K, Yan H, et al. Reanalysis of whole exome sequencing data in patients with epilepsy and intellectual disability/mental retardation. Gene 2019;700:168. Liu P, Meng L, Normand EA, et al. Reanalysis of clinical exome sequencing data. N Engl J Med 2019;380:2478. Ewans LJ, Schofield D, Shrestha R, et al. Whole-exome sequencing reanalysis at 12 months boosts diagnosis and is cost-effective when applied early in Mendelian disorders. Genet Med 2018;20:1564. Glazer AM, Wada Y, Li B, et al. High-throughput reclassification of SCN5A variants. Am J Hum Genet 2020;107:111. Zimowski J, Fidzánska E, Holding M, et al. Two mutations in one dystrophin gene. Neurol Neurochir Pol 2013;47:131. Yang J, Li SY, Li YQ, et al. MPLA-based genotype phenotype analysis in 1053 Chinese patients with DMD/BMD. BMC Med Genet 2013;14:29. Akiyama M, Titeux M, Sakai K, et al. DNA-based prenatal diagnosis of harlequin ichthyosis and characterization of ABCA12 mutation consequences. J Invest Dermatol 2007;127:568. Brandão P, Seco S, Loureiro T, et al. Prenatal sonographic diagnosis of Harlequin ichthyosis. J Clin Ultrasound 2019;47:228.

608 Genetic Disorders and the Fetus

400. Schorderet DF, Huber M, Laurini RN, et al. Prenatal diagnosis of lamellar ichthyosis by direct mutational analysis of the keratinocyte transglutaminase gene. Prenat Diagn 1997;17:483. 401. Bichakjian CK, Nair RP, Wu WW, et al. Prenatal exclusion of lamellar ichthyosis based on identification of two new mutations in the transglutaminase 1 gene. J Invest Dermatol 1998;110:179. 402. Akiyama M. Updated molecular genetics and pathogenesis of ichthyoses. Nagoya J Med Sci 2011; 73:79. 403. Akiyama M. ABCA12 mutations and autosomal recessive congenital ichthyosis: a review of genotype/phenotype correlations and of pathogenetic concepts. Hum Mutat 2010;31:1090. 404. Di Mario M, Ferrari A, Morales V, et al. Antenatal molecular diagnosis of X-linked ichthyosis by maternal serum screening for Down’s syndrome. Gynecol Obstet Invest 1998;45:277. 405. Aviram-Goldring A, Goldman B, Netanelov-Shapira I, et al. Deletion patterns of the STS gene and flanking sequences in Israeli X-linked ichthyosis patients and carriers: analysis by polymerase chain reaction and fluorescence in situ hybridization techniques. Int J Dermatol 2000;39:182. 406. Langlois S, Armstrong L, Gall K, et al. Steroid sulfatase deficiency and contiguous gene deletion syndrome amongst pregnant patients with low serum unconjugated estriols. Prenat Diagn 2009;29:966. 407. Muys J, Blaumeiser B, Jacquemyn Y, et al. The Belgian MicroArray Prenatal (BEMAPRE) database: A systematic nationwide repository of fetal genomic aberrations. Prenat Diagn 2018;38:1120. 408. Watanabe T, Fujimori K, Kato, K, et al. Prenatal diagnosis for placental steroid sulfatase deficiency with fluorescence in situ hybridization: a case of X-linked ichthyosis. J Obstet Gynaecol Res 2003;29:427. 409. Fassihi H, Liu L, Renwick PJ, et al. Development and successful clinical application of preimplantation genetic haplotyping for Herlitz junctional epidermolysis bullosa. Br J Dermatol 2010;162:1330. 410. Has C, Küsel J, Reimer A, et al. The position of targeted next-generation sequencing in epidermolysis bullosa diagnosis. Acta Derm Venereol 2018;98:437. 411. Dommering CJ, Mol BM, Moll AC, et al. RB1 mutation spectrum in a comprehensive nationwide cohort of retinoblastoma patients. J Med Genet 2014;51:366. 412. Castéra L, Gauthier-Villars M, Dehainault C, et al. Mosaicism in clinical practice exemplified by prenatal diagnosis in retinoblastoma. Prenat Diagn 2011;31:1106.

413. Lau CS, Choy KW, Fan DS, et al. Prenatal screening for retinoblastoma in Hong Kong. Hong Kong Med J 2008;14:391. 414. Kataoka A, Hirakawa S, Iwamoto M, et al. Prenatal diagnosis of a case of partial monosomy/monosomy 13 mosaicism: 46,XX,r(13)(p11q33)/45,XX,-13 suspected by nuchal fold translucency increasing. Kurume Med J 2011;58:127. 415. Julian-Reynier C, Chabal F, Frebourg T, et al. Professionals assess the acceptability of preimplantation genetic diagnosis and prenatal diagnosis for managing inherited predisposition to cancer. J Clin Oncol 2009;27:4475. 416. Drusedau M, Dreesen JC, Derks-Smeets I, et al. PGD for hereditary breast and ovarian cancer: the route to universal tests for BRCA1 and BRCA2 mutation carriers. Eur J Hum Genet 2013;21:1361. 417. Poulton A, Lewis S, Hui L, et al. Prenatal and preimplantation genetic diagnosis for single gene disorders: a population-based study from 1977 to 2016. Prenat Diagn 2018;38:904. 418. Clancy T. A clinical perspective on ethical arguments around prenatal diagnosis and preimplantation genetic diagnosis for later onset inherited cancer predispositions. Fam Cancer 2010;9:9. 419. Claes K, Dahan K, Tejpar S, et al. The genetics of familial adenomatous polyposis (FAP) and MutYH-associated polyposis (MAP). Acta Gastroenterol Belg 2011;74:421. 420. Douma KF, Aaronson NK, Vasen HF, et al. Attitudes toward genetic testing in childhood and reproductive decision-making for familial adenomatous polyposis. Eur J Hum Genet 2010;18:186. 421. Schaefer AM, McFarland R, Blakely EL, et al. Prevalence of mitochondrial DNA disease in adults. Ann Neurol 2008;63:35. 422. Gorman GS, Chinnery PF, DiMauro S, et al. Mitochondrial diseases. Nat Rev Dis Primers 2016;2:16080. 423. Frazier AE, Thorburn DR, Compton AG. Mitochondrial energy generation disorders: genes, mechanisms, and clues to pathology. J Biol Chem 2019;294:5386. 424. Djouadi F, Bastin J. Mitochondrial genetic disorders: cell signaling and pharmacological therapies. Cells 2019;8:289. 425. Schwartz M, Vissing J. Paternal inheritance of mitochondrial DNA. N Engl J Med 2002;347:576. 426. Luo S, Valencia CA, Zhang J, et al. Biparental inheritance of mitochondrial DNA in humans. Proc Natl Acad Sci U S A 2018;115:13039. 427. Rius R, Cowley MJ, Riley L, et al. Biparental inheritance of mitochondrial DNA in humans is not a common phenomenon. Genet Med 2019;21:2823.

CHAPTER 14

428. Marchington DR, Scott-Brown M, Barlow DH, et al. Mosaicism for mitochondrial DNA polymorphic variants in placenta has implications for the feasibility of prenatal diagnosis in mtDNA diseases. Eur J Hum Genet 2006;14:816. 429. Vachin P, Adda-Herzog E, Chalouhi G, et al. Segregation of mitochondrial DNA mutations in the human placenta: implication for prenatal diagnosis of mtDNA disorders. J Med Genet 2018;55:131. 430. Nesbitt V, Alston CL, Blakely EL, et al. A national perspective on prenatal testing for mitochondrial disease. Eur J Hum Genet 2014;22:1255. 431. González MDM, Ramos A, Aluja MP, et al. Sensitivity of mitochondrial DNA heteroplasmy detection using next generation sequencing. Mitochondrion 2020;50:93. 432. Kim MY, Cho S, Lee JH, et al. Detection of innate and artificial mitochondrial DNA heteroplasmy by massively parallel sequencing: considerations for analysis. J Korean Med Sci 2018;33:e337. 433. Rong E, Wang H, Hao S, et al. Heteroplasmy detection of mitochondrial DNA A3243G mutation using quantitative real-time PCR assay based on TaqMan-MGB probes. Biomed Res Int 2018;2018:1286480. 434. Harding AE, Holt IJ, Sweeney MG, et al. Prenatal diagnosis of mitochondrial DNA 8993T-G disease. Am J Hum Genet 1992;50:629. 435. Ferlin T, Landrieu P, Rambaud C, et al. Segregation of the G8993 mutant mitochondrial DNA through generations and the embryonic tissues in a family at risk of Leigh syndrome. J Pediatr 1997;131:447. 436. White SL, Shanske S, Biros I, et al. Two cases of prenatal analysis for the pathologic T to G substitution at nucleotide 8993 in mitochondrial DNA. Prenat Diagn 1999;19:1165. 437. Bartley J, Senadheera D, Park P, et al. Prenatal diagnosis of T8993G mitochondrial DNA point mutation in amniocytes by heteroplasmy detection. Am J Hum Genet 1996;59:A317. 438. Leshinsky-Silver E, Perach M, Basilevsky E, et al. Prenatal exclusion of Leigh syndrome due to T8993C mutation in the mitochondrial DNA. Prenat Diagn 2003;23:31. 439. Bouchet C, Steffann J, Corcos J, et al. Prenatal diagnosis of myopathy, encephalopathy, lactic acidosis, and stroke-like syndrome: contribution to understanding mitochondrial DNA segregation during human embryofetal development. J Med Genet 2006;43:788. 440. Amato P, Tachibana M, Sparman M, et al. Three-parent in vitro fertilization: gene replacement for the prevention of inherited mitochondrial diseases. Fertil Steril 2014;101:31.

Molecular Genetics and Prenatal Diagnosis 609

441. Costa-Borges N, Spath K, Miguel-Escalada I, et al. Maternal spindle transfer overcomes embryo developmental arrest caused by ooplasmic defects in mice. Elife. 2020;9:e48591. 442. Mobarak H, Heidarpour M, Tsai PJ, et al. Autologous mitochondrial microinjection; a strategy to improve the oocyte quality and subsequent reproductive outcome during aging. Cell Biosci 2019;9:95. 443. Zhang J, Liu H, Luo S, et al. Live birth derived from oocyte spindle transfer to prevent mitochondrial disease [published correction appears in Reprod Biomed Online. 2017 Jul;35:49. Konstandinidis, Michalis [corrected to Konstantinidis, Michalis]] [published correction appears in Reprod Biomed Online 2017;35:750]. Reprod Biomed Online 2017;34:361. 444. Green RC, Berg JS, Grody WW, et al. ACMG recommendations for reporting of incidental findings in clinical exome and genome sequencing. Genet Med 2013;15:565. 445. ACMG policy statement: updated recommendations regarding analysis and reporting of secondary findings in clinical genome-scale sequencing. Genet Med 2015;17:68. 446. Kalia SS, Adelman K, Bale SJ, et al. Recommendations for reporting of secondary findings in clinical exome and genome sequencing, 2016 update (ACMG SF v2.0): a policy statement of the American College of Medical Genetics and Genomics [published correction appears in Genet Med. 2017 Apr;19:484]. Genet Med 2017;19:249. 447. Haer-Wigman L, van der Schoot V, Feenstra I, et al. 1 in 38 individuals at risk of a dominant medically actionable disease. Eur J Hum Genet 2019;27:325. 448. Olfson E, Cottrell CE, Davidson NO, et al. Identification of medically actionable secondary findings in the 1000 genomes. PLoS One 2015;10:e0135193. 449. Amendola LM, Dorschner MO, Robertson PD, et al. Actionable exomic incidental findings in 6503 participants: challenges of variant classification. Genome Res 2015;25:305. 450. Hart MR, Biesecker BB, Blout CL, et al. Secondary findings from clinical genomic sequencing: prevalence, patient perspectives, family history assessment, and health-care costs from a multisite study [published correction appears in Genet Med 2019 Jan 22]. Genet Med 2019;21:1100. 451. Schwartz MLB, McCormick CZ, Lazzeri AL, et al. A model for genome-first care: returning secondary genomic findings to participants and their healthcare providers in a large research cohort. Am J Hum Genet 2018;103:328.

610 Genetic Disorders and the Fetus

452. Katz AE, Nussbaum RL, Solomon BD, et al. Management of secondary genomic findings. Am J Hum Genet 2020;107:3. 453. American Society of Human Genetics Board of Directors and American College of Medical Genetics Board of Directors. Points to consider: ethical, legal, and psychosocial implications of genetic testing in children and adolescents. Am J Hum Genet 1995;57:1233. 454. Committee on Bioethics. Ethical and policy issues in genetic testing and screening of children. Pediatrics 2013;131:620. 455. Ross LF, Saal HM, David KL, et al. Technical report: ethical and policy issues in genetic testing and screening of children. Genet Med 2013;15:234. 456. Kalynchuk EJ, Althouse A, Parker LS, et al. Prenatal whole-exome sequencing: parental attitudes. Prenat Diagn 2015;35:1030. 457. Wou K, Weitz T, McCormack C, et al. Parental perceptions of prenatal whole exome sequencing (PPPWES) study. Prenat Diagn 2018;38:801.

458. Brew CE, Castro BA, Pan V, et al. Genetics professionals’ attitudes toward prenatal exome sequencing. J Genet Couns 2019;28:229. 459. Horn R, Parker M. Health professionals’ and researchers’ perspectives on prenatal whole genome and exome sequencing: ‘We can’t shut the door now, the genie’s out, we need to refine it’. PLoS One 2018;13:e0204158. 460. Dondorp WJ, Page-Christiaens GC, de Wert GM. Genomic futures of prenatal screening: ethical reflection. Clin Genet 2016;89:531. 461. Harris S, Gilmore K, Hardisty E, et al. Ethical and counseling challenges in prenatal exome sequencing. Prenat Diagn 2018;38:897. 462. Richardson A, Ormond KE. Ethical considerations in prenatal testing: Genomic testing and medical uncertainty. Semin Fetal Neonatal Med 2018;23:1. 463. Horn R, Parker M. Opening Pandora’s box? Ethical issues in prenatal whole genome and exome sequencing. Prenat Diagn 2018;38:20.

15

Prenatal Diagnosis of Cystic Fibrosis Wayne W. Grody UCLA School of Medicine and UCLA Medical Center, Los Angeles, CA, USA

For many practitioners in the prenatal setting, cystic fibrosis (CF) represents their first exposure to a “molecular disease” – one that is screened at the DNA level to reveal risks to the fetus that are otherwise not apparent. Indeed, since heterozygous carriers of recessive mutations for the disease are entirely normal, as are the fetuses in most cases, prenatal screening and diagnosis for CF was not even feasible until the discovery of the causative gene in 1989. Unlike the trisomies and neural tube defects that obstetricians could visualize by ultrasound and screen for using biochemical signs of the disorders (maternal α-fetoprotein and other serum markers), CF is, except in the minority of cases with meconium ileus/echogenic bowel, an invisible disorder to the obstetrician and one that moreover had formerly been exclusively within the purview of pediatricians. Thus, the advent of population-based carrier screening for CF mutations in the prenatal setting was a true paradigm shift for obstetricians at the time, although, as we now know, it was merely the harbinger of even more sophisticated DNA-based tests to come.

Genetics and epidemiology CF is often considered the most common lifethreatening autosomal recessive disorder in North America. It is clearly inherited in an autosomal recessive fashion, in which the parents of an affected child are both obligate carriers. Though

physiologically entirely normal themselves, the at-risk couple has a 1-in-4 chance with each pregnancy of having a child with CF. The disorder is most common among Caucasians of European ancestry (including Ashkenazi Jews), in whom the birth incidence is about 1 in 3,500. The prevalence of the disorder is about 30,000 in the United States and 60,000–70,000 worldwide.1, 2 Likewise, the carrier rate is highest among non-Hispanic Caucasians at 1 in 25, but also found at appreciable levels in other ethnic and racial groups (Table 15.1). It is because of this panethnic carrier rate that universal population carrier screening was deemed useful in order to identify couples at risk who otherwise would only learn of their carrier status through the birth of an affected child.

Clinical features Although usually thought of as a pulmonary disease, CF actually affects many different organs and tissues. What most of these features have in common is abnormal ion transport and abnormally viscous or salty secretions. In the classic form of the disease, these viscous secretions in the lung are not readily cleared by the usual mechanisms and become culture media for bacteria that, over the years, become resistant to the available antibiotics. The most notorious species in this regard is Pseudomonas aeruginosa, and many patients ultimately

Genetic Disorders and the Fetus: Diagnosis, Prevention, and Treatment, Eighth Edition. Edited by Aubrey Milunsky and Jeff M. Milunsky. © 2021 Aubrey Milunsky and Jeff M. Milunsky. Published 2021 by John Wiley & Sons Ltd.

611

612 Genetic Disorders and the Fetus

Table 15.1 Cystic fibrosis mutation carrier frequencies and detection rates in various ethnic/racial groups Percentage

Approximate

of Group

carrier risk

mutations

Carrier

detected

frequency screening

after negative

African American

64

1/61

1/170

Ashkenazi Jewish

94

1/24

1/380

Asian American

49

1/94

1/180

Caucasian

88

1/25

1/200

72

1/58

1/200

Diagnosis

(non-Hispanic) Hispanic

fallopian tube secretions, but this is nowhere near as dramatic as in the male patients.3, 4 In the sweat glands, abnormal ion transport results in elevated levels of sodium and chloride secreted in the sweat. This is the basis of the well-known sweat test for diagnosis of CF5 and, in earlier days, clinicians licking the skin of suspected patients to assess the salty taste.

Source: Adapted from American College of Medical Genetics and Genomics, Technical Standards and Guidelines for CFTR Mutation Testing, 2006. Available at http://www.acmg.net

die from resistant infections by this organism or from the chronic lung damage it causes. Up until that time, they exhibit features of chronic obstructive pulmonary disease, with coughing, bronchitis and bronchiectasis, hypoxia, and recurrent pneumonias that often require admission to the hospital for treatment. In the gastrointestinal tract, viscous secretions can result in meconium ileus and intestinal obstruction. This affects under 10 percent of CF patients and is an emergency in the newborn period. In about 85 percent, the pancreatic ducts become obstructed, resulting in chronic malabsorption of fats and proteins and leading to failure to thrive in infants and young children. In the reproductive tract, almost all males with CF exhibit a congenital malformation called congenital bilateral absence of the vas deferens (CBAVD), which causes obstructive azoospermia and infertility. Of note, sperm production is normal, and these men can father children through the procedure of sperm aspiration. Conversely, some patients with atypical CF may experience only the infertility and be identified when they present to infertility clinics and are tested for CFTR mutations (which is now part of the standard workup for male infertility). (The CFTR genotypes associated with isolated CBAVD are quite complex and are discussed in detail later in this chapter.) There are some reports of compromised fertility in female CF patients, presumed due to viscous cervical or

Until the identification of the gene in 1989, diagnosis of CF in affected individuals relied primarily on clinical history with attention to the features described above. Family history is also contributory in those cases where there is a previously affected sibling or other relative, but more often the proband is the first case in the family. Two biochemical measurements are also used. For newborn screening, which is now offered in all 50 states and many other countries, measurement of immunoreactive trypsinogen (IRT) in the serum can be used as an early sign of pancreatic obstruction; this is in every sense a screening test, not a diagnostic one, and false-positive and false-negative results are common.6 IRT is elevated in neonates with early pancreatic changes; once full-blown pancreatic insufficiency develops, the IRT level falls below normal. For postnatal diagnosis, the sweat test, or sweat chloride test, has been the mainstay of clinical chemistry diagnosis of CF for many decades. Chloride concentrations above 60 mmol/L are generally considered diagnostic. Sweat chloride levels generally trend with the more classic or severe mutations, but there are exceptions and it is possible to have classic lung disease with normal sweat chloride, or vice versa. The sweat test requires skill and experience to perform properly, so false results are possible; also, it is said to be less reliable in the immediate newborn period.5 Lastly, diagnosis can be made at the molecular level by the observation of two mutations in the CFTR gene. Yet there are patients with undisputed clinical CF in whom only a single mutation is found, even by complete gene sequencing. It is assumed that such cases involve a second mutation in a noncoding region of the gene (not addressed by the sequencing test).

CHAPTER 15

Treatment Although there is as yet no cure for CF, advances in supportive therapies have dramatically increased the life expectancy, with median survival progressing from under 1 year 80 years ago to the mid-40s today. Treatments consist of everything from basic physical therapy to clear lung secretions to specific mutation-targeted molecular therapies. The viscosity of these secretions can be reduced for easier clearance by the administration of deoxyribonuclease-𝛼 (Pulmozyme , Genentech).7 Clearly a major credit for the increased lifespan is owed to the ever-expanding arsenal of antibiotics for treatment of new and resistant infections. The availability of pancreatic enzyme supplements addresses the problems of malabsorption, malnutrition, and failure to thrive. Steroidal and nonsteroidal anti-inflammatory drugs improve outcome by reducing reactive damage from the infections. For patients who eventually reach end-stage lung disease, lung transplant has proved a savior, assuming the patient survives the surgery and immunosuppression. For patients with concomitant cardiac disease secondary to pulmonary hypertension, combined heart–lung transplants have proven effective.8 Of course, the ultimate dream in CF management is correction of the basic defect by gene replacement therapy. Alas, despite a great deal of work in this area, significant hurdles remain in the form of adequate gene delivery to the target tissue (bronchial epithelium) and host response against the commonly used viral vectors. More recently, the advent of gene editing for mutation correction has offered promise, though again there are many technical challenges to overcome.9 In the interim, however, there have been exciting advances in what might be called the next best thing to gene therapy: direct targeting of specific mutations by small-molecule drugs, analogous to what is being done in oncology. These drugs are collectively called “CFTR modulators;” they represent an elegant example of “personalized medicine,” in that the treatment is tailored to the patient’s specific, personal mutation(s).10 The first such drug to emerge from the research phase and come on the market was ivacaftor (Kalydeco , Vertex Pharmaceuticals), which specifically targets the G551D mutation that is found in about 5

®

®

Prenatal Diagnosis of Cystic Fibrosis

613

percent of CF patients.11 The drug improves CFTR ion transport and therefore is called a “CFTR potentiator.” Other drugs have since emerged that target more common mutations, including the predominant mutation ΔF508, so that at present a triple-drug cocktail can be effective in about 90 percent of CF patients.12 It is worth considering the advent of these targeted therapies in the context of this chapter, since carrier screening is predicated on the fact that there is no cure for CF, yet these drugs would seem to put that goal within reach. The obvious question is whether couples will be less inclined to terminate affected pregnancies when the fetus carries one of these “targetable” mutations.13

Discovery of the cystic fibrosis gene The causative gene for CF was identified in 1989, which predates by 1 year the launch of the Human Genome Project. Thus, this gene, the target of an intense search for many years, was isolated the old-fashioned way, by linkage analysis within families followed by successive cloning, DNA “walking,” and DNA “jumping” to narrow down the region on chromosome 7, and finally the characterization of candidate genes within the region to see which one seemed to fit the features of CF based on tissue-specific gene expression. The identified gene, expressed in lung, nasal epithelium, sweat glands, colon and liver,14 was found to carry a mutation in the few subjects analyzed (the famous ΔF508 mutation) and thus fit the requirements. Like dystrophin before it, the protein product was named after the disease, which is unfortunate since the protein did not evolve merely to cause cystic fibrosis. Nevertheless, we are left with the ungainly name of cystic fibrosis transmembrane conductance regulator, with the gene name being CFTR. The CFTR gene and its protein product

The CFTR gene is relatively large, spanning about 250 kb and containing 27 exons.15 It is located on the long arm of chromosome 7 at 7q31.2. The CFTR protein contains 1,480 amino acid residues and has a molecular weight of 168 kDa. Its shared homology with known cell membrane transport proteins gave the first clue that it functions as

614 Genetic Disorders and the Fetus

an ion channel. Further study revealed it to be an active chloride transporter, dependent upon ATP for energy. The protein itself consists of both membrane-spanning domains (the pores) and a nucleotide-binding domain where ATP binds and is hydrolyzed.16 This deduced function was satisfying in that it could explain many of the symptoms and features of cystic fibrosis. CFTR mutations and variants

As just noted, the first few patients studied at the time of the gene discovery all had a specific three-nucleotide deletion of codon 508, designated ΔF508 (also referred to, by purists, as c.1521_1523delCTT using the formal Human Genome Variation Society nomenclature).17 This finding prompted early hopes that a CF carrier screening test would be simple and easily implemented. However, it was not long before those hopes were dashed, as a great number of additional mutations began to be catalogued, many of them extremely rare or confined to single families (so-called “private” mutations). While ΔF508 accounts for about 70 percent of Caucasian carriers of northern European ancestry, the next most common mutation in that population, G542X, is found in only about 1.5 percent of carriers, and after the next few mutations the carrier frequency drops to well under 1 percent. Thus, a screening test for CFTR carrier mutations was not to come easily. Indeed, we now know that the mutational spectrum of the gene is extremely heterogeneous, with close to 2,000 mutations catalogued thus far (available at the Cystic Fibrosis Mutation Database: http://www.genet.sickkids.on.ca). They are spread across all the exons of the gene, with no particular “hotspots.”18 At time of this writing, 39 percent of the mutations are missense, 16 percent are frameshift, 11 percent splicing defects, 8 percent nonsense, and about 5 percent are insertions or deletions. It is largely because of this mutational heterogeneity that population carrier screening for

CF mutations took 12 years to be launched from the time the gene was discovered. Though not necessarily cited as such in CFTR testing reports, some laboratorians and practitioners have found it useful to delineate classes of mutations based on their effect on the protein product of the gene. There are five classes (Table 15.2), as follows: class I mutations are the most severe, resulting in truncation of the protein product, as caused by nonsense mutations or splice variants leading to exon skipping (e.g. the nonsense mutation G542X and the splice-junction mutation 1717–1G→A); class II mutations result in proteins that are intrinsically functional but are not processed properly after translation and never reach the cell membrane (e.g. the microdeletion mutation ⊗F508); class III mutations result in proteins that do not properly bind or hydrolyze ATP (e.g. the missense mutation G551D); class IV mutations result in proteins that do bind ATP but are inefficient in chloride transport (e.g. the missense mutation R117H); and class V mutations result in decreased levels of normally functioning proteins, usually due to alternative splicing (e.g. 3849+10kb C→T).19 Again, although most clinical laboratory reports will not specify the mutational result by classes, they are worth keeping in mind for their mechanistic implications as we move further into the era of mutation-specific therapies (discussed earlier).11, 12

Genotype–phenotype correlation The spectrum of severity of clinical symptoms in CF is extraordinarily broad, ranging from infants who die in the newborn period of meconium ileus to middle-aged adults who experience only chronic sinusitis and have no idea that they carry two CFTR mutations. It goes without saying that mutations associated with the former, along with serious lung disease and pancreatic insufficiency, are classified as “severe,” while those associated

Table 15.2 Classes of CFTR mutations Class

I

II

III

IV

V

Defect

Protein truncation

Post-translational processing

ATP-binding

Cl− transport

Decreased product

Examples

G542X; 1717–1G→A

ΔF508

G551D

R117H

3849+10kb C→T

CHAPTER 15

with the latter “atypical” forms are classified as “mild.” Pancreatic insufficiency to a degree requiring pancreatic enzyme supplementation (seen in 85 percent of CF patients) tends to track with the “severe” mutations.20, 21 Unfortunately, however, those designations are not absolutely predictive: some patients with so-called “mild” mutations may have classical disease replete with pancreatic insufficiency, while there are homozygotes for the most classic “severe” mutation, ΔF508, who experience only mild or subclinical lung disease well into adult life.22–25 Variable expressivity even extends to the biochemical abnormalities, in that a patient may (though rarely) have classic lung disease but normal or near-normal sweat chloride levels.26, 27 The reason, clearly, is that the expression of all of these mutations is subject to the action of various known and unknown modifier genes and other host factors (see later). But aside from academic interest, this relatively poor genotype–phenotype correlation makes genetic counseling for the results of prenatal and postnatal diagnostic tests fraught with uncertainty. If a prenatal mutation test result is poorly predictive as to expected mild or severe disease, how are parents expected to make an informed choice regarding continuation or termination of the pregnancy?

Congenital bilateral absence of the vas deferens Perhaps the most striking example of clinical variability in CF involves the feature of male infertility due to CBAVD. This malformation is seen in almost all males with CF but, surprisingly, it can also be seen as an isolated feature in the absence of any other signs of CF, associated with certain CFTR mutations and variants.28 Most patients with isolated CBAVD have a classical CF mutation on one allele and a more “mild” or atypical variant on the other allele. It may be that the male reproductive apparatus requires higher levels of CFTR activity for proper development than do the lungs and pancreas for proper function, so a partial compromise not dramatic enough to cause classical CF symptoms can still cause CBAVD.29 This has led to some debate over the primary diagnosis in such individuals. Do they have an extremely mild/atypical form of CF or do they in fact have an entirely different disease?30, 31 (Examples of other genes causing two

Prenatal Diagnosis of Cystic Fibrosis

615

different diseases are not unknown in medical genetics.) Whatever the nomenclature, it is now widely accepted that otherwise healthy adult males presenting to infertility clinics should be checked for CBAVD and then for CFTR mutations. The two mutations most commonly associated with CBAVD are R117H and 3849+10kb C→T, with the former being by far the more common.28, 32 Both mutations are influenced in their expressivity and penetrance by a tandem repeat polymorphism in intron 8 of the CFTR gene consisting entirely of thymidines. The alleles commonly seen in the population are runs of 5, 7, or 9 thymidines (designated 5T, 7T, and 9T, respectively). The repeat is located toward the 3′ -end of intron 8, near the splice-acceptor site, and the 7T and 5T alleles result in reduced RNA splicing efficiency and hence gene expression (with 5T worse than 7T). The details of the interaction are rather complex (Table 15.3) and depend upon whether the 5T or 7T variants are in cis or trans (i.e. on the same or opposite chromosome) with a variable mutation such as the two noted above.33, 34 In general, having 5T in cis with a mutation like R117H (and in the presence of another CF mutation on the opposite allele) causes enough of a decrease in gene expression to result in CF symptoms, albeit often fairly mild, whereas 5T in trans with R117H is more likely to result in CBAVD (if anything). Likewise, 7T in cis with R117H is more likely to result in CBAVD (or nothing at all). 7T in trans with R117H has no effect as long as the mutation has the “normal” 9T variant in cis. Lastly, homozygosity for 5T, even in the absence of any exonic CFTR mutation, is associated with CBAVD.35 And to make matters even more complicated, the effect of the 5T variant is further influenced by another tandem repeat within intron

Table 15.3 R117H/polyT genotype–phenotype correlations Genotype

Phenotype includes

R117H-5T/CF mutation

Nonclassic CF, PS CF

R117H-7T/CF mutation

Asymptomatic female, CBAVD,

5T/CF mutation

Asymptomatic, nonclassic CF,

nonclassic CF, PS CF (including R117H) 7T or 9T/CF mutation

CBAVD Asymptomatic

CF, cystic fibrosis; PS, pancreatic sufficiency; CBAVD, congenital bilateral absence of the vas deferens.

616 Genetic Disorders and the Fetus

8, a polyTG tract adjacent to the polyT tract. Eleven TG repeats counters the adverse function of the 5T, whereas 12 or 13 TG repeats are associated with the abnormal phenotypes.36 At present, this is largely of academic interest, as measurement of the TG tract is not routinely performed by clinical laboratories, unlike measurement of the polyT tract.

Modifier genes The variable penetrance and expressivity of CFTR mutations in different patients has called into question whether CF is truly a “single-gene disorder,” as it has in many other Mendelian conditions. This variance must be caused by something extraneous to the CFTR gene itself, whether environmental, epigenetic, or genetic (and most likely a combination of all three). It is well known that environmental factors such as cigarette smoke, exposure to infectious agents, general nutrition level, socioeconomic status, and compliance with antibiotic and physical therapies may adversely affect outcomes in patients with identical mutations.29, 37 But there are clearly genetic host factors as well. Just as the intron 8 polyT tract can affect clinical expressivity, so can a variety of other genes, both known and unknown. Much of the work in this area is in its infancy, but variants in a number of genes and putative regulatory regions appear to impact severity or prevalence of lung, pancreatic, or gastrointestinal manifestations.29, 38 However, as most of these findings are the products of genome-wide association studies (GWAS), they are as yet not sufficiently predictive for practical use in CF genetic testing. As genome-level DNA sequencing (whole-exome or whole-genome) becomes more routine, it is expected that many of these factors will be further defined in CF patients, hopefully allowing for improved prognostication, surveillance, and prevention of complications.

Ethnic variation in mutation frequencies As noted, the vast majority of the approximately 2,000 mutations identified in the CFTR gene are extremely rare or even “private.” Of the recurrent mutations, ΔF508 is by far the most common, but its frequency, along with those of many

others, varies markedly among different ethnic and racial populations. Worldwide, it accounts for 68 percent of CF chromosomes (at least of those tested);1 it is at higher frequency in individuals of northern European ancestry. In contrast, it accounts for only about 50 percent of African American carriers39 and Hispanic Americans.40 In Ashkenazi Jews, some of the ΔF508 frequency is supplanted by a mutation unique to this population, W1282X,41 while in African Americans the second most-common mutation is 3120+1 G→A. Beyond ΔF508, other recurring (though much less common) mutations detected in the general population include G542X, R553X, R334W, R117H, 3849+10kb C→T, G551D, 621+1 G→T, and N1303K. As CF is a rare disease among Asians, not much is known about common or unusual mutations in that racial group (and the same for Native Americans).40

Development and implementation of public policy for CF population carrier screening and the core mutation panel As is true for all single-gene disorders, prenatal diagnosis for CF cannot proceed unless and until the mutational genotypes of both parents are known; otherwise, one does not know what to look for in the fetus. But as a pure recessive disease, the majority of carriers do not know they are carriers, and the majority of at-risk couples do not know they are at risk until they give birth to their first affected child. That is the impetus for initiating population carrier screening, in order to identify carriers and couples at risk so that they may be offered prenatal diagnosis and the option to terminate an affected pregnancy and thereby avoid the birth of that first affected child. Of course, that is the same impetus for past successful carrier screening programs such as those for thalassemia in the Mediterranean region and for Tay–Sachs and other recessive diseases in the Ashkenazi Jewish population. The difference is that CF is not restricted or markedly concentrated within a single ethnic or racial population but rather is panethnic. That is the reason why CF was the first universal population carrier screen to be implemented, offered to all pregnant couples or to those planning

CHAPTER 15

a pregnancy. And, as noted at the start of this chapter, it was the first program to be instituted using molecular (DNA-based) technology. But how did we get to this place where universal CF carrier screening is the standard of care? It was a long and tortuous road, interrupted by speed bumps and controversies. It started when the CFTR gene was discovered in 1989. At that time a consortium representing the National Center for Human Genome Research (now the National Human Genome Research Institute [NHGRI]), the American College of Medical Genetics (now the American College of Medical Genetics and Genomics [ACMG]) and the American College of Obstetricians and Gynecologists (ACOG) decided that CF population carrier screening was worth considering but should not be initiated until a number of feasibility and ethical issues could be sorted out via pilot studies and expert committees (in fact, an actual moratorium was laid down until these things could be accomplished).42, 43 Of particular concern was the mutational heterogeneity of the gene and the fact that no screening test of reasonable cost (at that time) would be capable of picking up all carriers. Would such an imperfect test actually do more harm than good? Would it make many couples anxious without providing them with actionable information (for example, those couples in whom one partner tests positive and the other negative)? A number of pilot studies were funded under the sponsorship of the Ethical, Legal and Social Implications (ELSI) program of NHGRI. For several years these studies explored the practical feasibility, acceptance, and understanding of carrier screening in those with and without a family history of CF. The settings varied from large to medium-sized cities, from mostly Caucasian to multi-ethnic populations, from university-based to HMO clinics, and from pregnant couples to those not yet pregnant.44–47 All of the studies demonstrated feasibility of the screening programs, but the highest uptake (interest in being screened) occurred in the author’s own study,47 probably because it was set entirely within prenatal clinics and exclusively targeted couples who were pregnant. This is not surprising, as early carrier screening programs had demonstrated higher interest among couples who were already pregnant than in those not yet thinking about optimizing reproductive outcomes.

Prenatal Diagnosis of Cystic Fibrosis

617

In any case, these findings influenced the expert consensus panel that was convened at NIH in 1997 to evaluate the results of the pilot studies and ultimately recommended that CF carrier screening “be offered to all pregnant couples and those planning a pregnancy.”48 The expert panel did not address a number of practical implementation questions, leaving those to future expert groups. Such questions included: Should CF screening be offered only to those in the “higher risk” groups such as Caucasians and Ashkenazi Jews, or opened up to all individuals regardless of ethnicity? Should screening be offered in a sequential fashion – first to the pregnant woman and then to her reproductive partner only if she tests positive? How many of the 1,500 (at that time) known CFTR mutations should constitute a minimal core screening panel? Can practitioners go beyond the core panel if they or the patients wish? How should test results and residual risks be reported? These questions were indeed taken up by a steering committee and various subcommittees, with representatives from ACMG, ACOG, and NHGRI.49 As is now well known, it was decided that screening would be universal/panethnic, using a minimal core panel of the 25 most common and most phenotypically severe mutations (recognizing that genotype–phenotype correlations are far from perfect) (Table 15.4), based on data from the Cystic Fibrosis Foundation and large clinical reference laboratories; any mutation representing at least 0.1 percent of the mutant alleles in the affected CF patients within these databases was included.50 The resulting number, 25, was substantially greater than the number of mutations tested during the pilot study phase, which was initially just 6: ⊗F508, G542X, W1282X, N1303K, G551D, and R553X.47 Though the patient databases used were predominantly Caucasian, effort was made to include mutations unusually prevalent in other groups as long as they contributed at least 0.1 percent to the overall mixed population; thus, for example, the most common African mutation, 3120+1G→A, was included, as was the common Ashkenazi Jewish mutation, W1282X. Three years later, the core panel was modified with the removal of two variants: 1078delT and I148T:51 the former was discovered to be so rare that it was virtually never seen in screening and should not have reached the 0.1 percent threshold

618 Genetic Disorders and the Fetus

Table 15.4 The original American College of Medical Genetics and Genomics core mutation panel for population cystic fibrosis carrier screening50 ΔF508

ΔI507

G542X

G551D

W1282X

N1303K

R553X

621+1G>T

R117H

1717-1G>A

A455E

R560T

R1162X

G85E

R334W

R347P

711+1G>T

1898+1G>A

2184delA

1078delTa

3849+10kbC>T

2789+5G>A

3659delC

I148Ta

3120+1G>A a These

two mutations were removed in 2004.51

Source: Grody WW, Cutting GR, Klinger KW, et al. Laboratory standards and guidelines for population-based cystic fibrosis carrier screening. Genet Med 2001;3:149.50

set for inclusion, while the latter was discovered not to be a mutation at all but merely a benign variant (polymorphism); those CF patients who had I148T in the original affected cohort were later found to carry another, truly pathogenic mutation elsewhere in the gene on the same chromosome.52–54 So the revised core mutation panel for population screening was and remains 23, though there is now more flexibility in going well beyond that number (see “Expanded panels”). After careful consideration, it was decided that either the “couple” or “sequential” model of offering carrier screening could be used, as long as test results were given to both members of the couple; on that point it excluded one particular model in which couples testing positive–negative are reported as “negative” in order to spare them the resulting risk uncertainty.55, 56 It was felt to be unethical not to disclose a test result to a patient who had consented to it, and, moreover, in that model one loses the ancillary benefit of alerting other family members of the person who tests positive – the so-called “cascade” effect of genetic screening.57 In an Appendix to the recommendations, model test reports were provided covering various combinations of results, including relation of mutation R117H to the polyT tract, as well as a table showing the a priori carrier rates of various racial/ethnic populations and the residual risk of still being a carrier after testing negative with the core mutation panel (adapted here as Table 15.1).50 These risks are used by genetic counselors in the post-test counseling session. As shown, the test panel has the highest clinical sensitivity, 94 percent, in those of Ashkenazi Jewish descent (thanks largely to the combined prevalence of the ⊗F508 and W1282X

mutations), compared to 88 percent in the general non-Hispanic Caucasian population, and progressively less in the other ethnic/racial groups. Although the core panel was designed strictly for use in carrier screening in individuals with no family history of CF, many laboratories began to use the same assay for other purposes, such as diagnostic testing in symptomatic patients. One can do the math and figure out the sensitivity for detecting two mutations instead of one, which comes out to about 78 percent for the Caucasian population (about 21 percent of affected patients will show only a single mutation – which is supportive but not conclusive evidence for a diagnosis of CF – and about 1 percent will show no mutation). This should not be an issue in prenatal diagnosis, however, because one would not attempt to test the fetus without first knowing the identity of the mutations in both the father and mother. Since the R117H mutation exhibits different phenotypes depending on the length of the associated polyT tract in intron 8, the recommendations call for measuring the number of Ts – but only as a reflex test after R117H is identified in the person being screened. The reason for that approach is that the 5T allele is rather common in the general population and if both parents screened positive for the 5T, their baby, if male, would be at risk of CBAVD (homozygous 5T, even in the absence of any other CFTR mutation). Since the goal of this effort was to screen for risk of CF, not male infertility, there was a fear that too many 5T carriers, with no inherent risk of CF, would be identified in a first pass if everyone were screened for it. That is why it was designated a reflex test. In fact, since R117H even in cis with 5T is likely to produce mild disease, there was much debate

CHAPTER 15

about whether to include it at all in the core screening panel. In the end it was felt to be too common to completely ignore. On the other hand, a multinational European consensus group, considering the same question for their populations, decided against including R117H (and polyT).58 The committees fully recognized that this type of complex molecular screening for a disease previously in the domain of pediatricians and pulmonologists would represent a significant change for obstetricians who would be the primary providers of the test in the prenatal setting. In conjunction with ACMG, ACOG developed educational materials that were sent to all their members, and a “ramp-up” period of at least 6 months was specified in order to give laboratories and providers adequate time to incorporate this new paradigm into their practice. Indeed, laboratories faced their own unique challenge in suddenly having to test for 25 mutations in a multiplex type of assay when no commercial kits or reagents for doing so were available at the time. Fortunately, the law of supply-and-demand prevailed, as equipment and reagent vendors perceived a lively market once millions of couples were going to be screened; previously, diagnostic molecular genetics was a sort of backwater in the commercial world, compared to the much higher volume tests in molecular microbiology and molecular oncology. The result was a plethora of kits and platforms for purchase that were tailored to assaying precisely those 25 mutations (see “Laboratory methods”). In actual practice, it took something like 2–3 years for the majority of obstetricians to get on board, and even several years into the program there were still appreciable numbers of practitioners not routinely offering CF carrier screening to their patients.59, 60 Given that the procedure had been deemed standard of care by ACOG, the risk of liability from an affected child born to a couple who had not been offered screening is only too obvious.

Laboratory methods As stated, developing a multiplex assay for detection of 25 mutations is no simple task. Whereas detection of one or two mutations in a gene is relatively straightforward and is often accomplished using in-house, laboratory-developed methods, most laboratories had to depend on commercial

Prenatal Diagnosis of Cystic Fibrosis

619

vendors for the array-based and other assays designed to cover the ACMG core mutation panel. Laboratory directors soon had a diverse array of platforms to choose from, utilizing techniques such as microarrays, reverse-hybridization strips (also called dot blots), amplification-refractory mutation system (ARMS), oligonucleotide ligation assay (OLA), liquid bead arrays, fluorescence resonance energy transfer (FRET), and microbead suspension hybridization assays.61 The starting material for all of them is DNA which can be isolated from amniocytes or chorionic villus samples for prenatal testing, or from blood, saliva or buccal brushes for carrier screening. A summary of the technical advantages and disadvantages of the various methods is presented in Table 15.5. In general, all of them have produced robust accuracy in blinded proficiency tests,62, 63 and clinical practitioners need not concern themselves with the nuances, though they should strive to use only those laboratories that are CLIA- and College of American Pathologists (CAP)-certified.

Expanded panels Commercial device and reagent manufacturers are not the only ones who saw a large and appealing market in universal CF carrier screening. Commercial reference laboratories were also obliged to re-evaluate clinical molecular diagnostics as a potentially lucrative sector of the clinical laboratory market. But to capture larger market share, they needed to do something to distinguish themselves from their competitors, and the way they chose to do it was to boast of ever larger CFTR mutation panels. Despite the fact that the original ACMG recommendation strongly discouraged going beyond the 25 mutations in the core panel,50 the ink was hardly dry on that statement when commercial laboratories began publishing and advertising that they could offer larger panels with 40 or 70 or 100 or more mutations, allegedly promising greater sensitivity at detecting more carriers than the standard panel.64, 65 Considering that the carrier percentages of the additional mutations are all well under 0.1 percent, it seems hard to believe that their addition could noticeably increase clinical test sensitivity, but arguments were made that some of the additional mutations were specific to non-Hispanic/non-Caucasian ethnic

Table 15.5 Methods of mutation detection Method

Principle

Advantages

Disadvantages

Allele-specific oligonucleotide

Individual wild-type or mutant probes hybridize

Can be automated

Platform design and

hybridization (ASO)

to target (patient) DNA bound to membrane

Multiplex possible

interpretation of results can be complex

Reverse dot-blot hybridization

Probe pairs (wild-type and mutant) are bound to membrane and hybridized with target

Capable of high throughput Multiplex possible

Difficult to customize or add new mutations

(patient) DNA Rapid and robust assay Amplification-refractory mutation system (ARMS), also

PCR primers designed to amplify only a specific

Rapid and reliable

(typically mutant) sequence

Absence of product implies negative result if there is no

called allele-specific

paired wild-type reaction

amplification

Assays without paired wild-type reactions cannot differentiate between the heterozygous and homozygous mutant state

Oligonucleotide ligation assay (OLA)

Allele-specific PCR followed by ligation with probes to identify mutant and wild-type sequence

Liquid bead arrays

Multiplex PCR followed by hybridization to beads with covalently bound universal tags or allele-specific capture probes. A fluorochrome coupled to a reporter molecule

Capable of high throughput Software automatically analyzes data and

Detection requires use of automated DNA sequencer

creates summary reports Capable of high throughput Software analyzes data automatically and

Detection requires the use of specialized equipment

enables results for reflex polymorphisms to be revealed only as appropriate or on demand

quantifies hybridization Fluorescence resonance energy transfer (FRET)

Hybridization of patient DNA to a normal or mutant probe forms a structure that is

Capable of high throughput Rapid

recognized and cleaved by a proprietary enzyme. The released fragment hybridizes to

Software analyzes data automatically and

a cassette containing a reporter and

enables results for reflex polymorphisms to be

quencher molecule forming a second

revealed only as appropriate or on demand

structure, which is enzymatically cleaved, generating fluorescence signal

Detection requires the use of specialized equipment

Microarray hybridization

Hybridization of patient DNA to probes on a microarray (or chip)

Capable of high throughput Rapid Relatively easy to customize or add new

Detection requires the use of specialized equipment Expensive

mutations DNA sequencing (Sanger method)

Sequence individual regions, exons or complete CFTR gene

Can theoretically identify all mutations within the amplicons

Relatively expensive Relatively slow Cannot identify large deletions Likelihood of revealing variants of uncertain clinical significance (VUS)

DNA sequencing (next-generation method)

Sequence individual regions, exons or complete CFTR gene

(NGS)

High throughput Allows for integration of CF screening with

Requires very expensive equipment

expanded carrier screening for many other

Limited ability to identify large

disorders

deletions Likelihood of revealing variants of uncertain clinical significance (VUS)

Mass spectrometry/time-of-flight

Primer extension to detect a specific mutation

Rapid

Cannot detect large deletions

High resolution

Only detects known mutations

Multiplex possible

Requires expensive, specialized

Robust Automated

equipment

622 Genetic Disorders and the Fetus

groups that had been overlooked by the original Cystic Fibrosis Foundation cohort, which consisted mainly of northern European Caucasians.66, 67 On the other hand, some of these panels included variants of uncertain pathogenicity, such as D1270N, D1252, G662D, L997F, and R117C.68–70 Such mutations were deliberately not included in the ACMG panel so as not to present parents with ambiguous decision making. Some suggested that the competitive quest for ever larger CFTR screening panels at that time was not only unseemly but also unscientific, as summarized in a commentary by this author and colleagues.71 The selection of additional mutations was in some sense arbitrary, and because they were so rare, very little was known about them, so how could one be sure of their true penetrance and pathogenicity? Remember that the original ACMG core panel contained a serious error – the inclusion of the I148T variant which turned out to have no pathogenicity whatsoever – despite almost 3 years of careful vetting; this experience no doubt haunted the original committees and produced a certain hesitancy in dramatically expanding the mutation screening panel. How many other “I148Ts” may be lurking among the expanded panels currently offered? Nevertheless, it must be recognized that much has changed in the last 20 years in both molecular technologies and our understanding of the many variants in the CFTR gene. With the cost of DNA sequencing ever-diminishing since the introduction of next-generation sequencing (NGS), one may ask why not begin screening for CF carriers by that technique, enabling the easy detection of either a larger panel of targeted mutations or indeed any and all variants in the coding regions of the CFTR gene? DNA sequencing, even by the slower Sanger method, has long been accepted for postnatal CF diagnosis, especially for those patients with atypical or ambiguous clinical features and inconclusive targeted mutation tests. The resistance to its implementation in carrier screening and prenatal diagnosis has been because of the risk that it will pick up all sorts of single-nucleotide changes (missense variants) that have not been seen or reported before, with nothing known about their pathogenicity or lack thereof, and found in a parent or fetus with no phenotype to give us any clues. In the

sequencing world, these are the notorious “variants of uncertain significance” (VUS), and they could potentially place a pregnant couple in the untenable position of trying to make an irreversible reproductive decision based on little or no predictive information. The first move in this direction was the clearance by the US Food and Drug Administration (FDA), in 2013, of a targeted 139-variant CFTR panel on the Illumina MiSeq, a popular NGS instrument. In fact, this was the first FDA-approved NGS assay of any type in laboratory medicine.72 Importantly, these variants were not arbitrarily selected, as in so many previous expanded panels, but are based on a sophisticated study of all 2,000 CFTR mutations for prevalence (>0.01 percent of the total), phenotype in patients, and molecular and functional studies in vitro, the results of which are accessible in a database called CFTR2 (https://cftr2.org/). After a process of elimination, the investigators were left with these specific mutation targets that were concluded to be disease-causing.73 In light of these developments, along with widely accepted criteria (also promulgated by ACMG) for objective classification of novel sequence variants,74 ACMG has revisited the initial CF carrier screening recommendations.75 While stopping short of abandoning the original minimum screening panel (now affectionately referred to as “the ACMG-23”), the new guideline leaves open to laboratory discretion the use of larger panels or even complete gene sequencing. In part this was a recognition of the contemporary state of genetic testing technology, but also a recognition that “the cat is out of the bag” and more comprehensive gene and genomic sequencing is already here to stay and being applied to both diagnostic and screening indications. One reflection of this state is the heavy marketing and popular uptake of highly expanded carrier screening panels encompassing as many as 300 or more diseases, again made possible by the high-throughput nature of NGS. Aside from covering a great many genes, most of these offerings utilize comprehensive exome sequencing, essentially delivering an unlimited mutation screening panel for CF and all the other diseases as well. However, all the concerns raised above regarding the handling and reporting of VUSs and prediction of likely disease severity apply here as well.76

CHAPTER 15

Outcomes of the CF carrier screening program At time of this writing, it has now been almost 20 years since the official launch of population-based CF carrier screening in the United States. Even allowing for some delay in the ramping-up period, that should have been enough time to assess its outcomes. Strangely, however, hard outcomes data are difficult to come by, as the US healthcare system is so fragmented and no registry is kept of CF screening results. The best early data came out of northern California, where the incidence of CF births appeared to have decreased by about 50 percent compared to the prescreening days.77 A more recent meta-analysis found a somewhat broader range in various cited studies.78 It appears that much of the residual birth incidence can be ascribed to couples who consented to the carrier screening procedure but then got withdrew, either at the prenatal diagnosis or pregnancy termination step and decided to go on with the pregnancy regardless (D. Witt, personal communication). This is understandable for a disease like CF, which exhibits no mental retardation or major physical malformations. In addition, as would also be expected, the concurrent presence of a fetal abnormality on ultrasound (such as echogenic bowel; see later) influenced the choice for pregnancy termination. Given that the lifetime cost of medical care for a patient with CF living to the median life expectancy is US$1–2 million, one can draw a general conclusion that the carrier screening program has been cost effective.

Special prenatal diagnosis situations Despite the complexity of CFTR mutations, the majority of pregnant couples undergoing this screening will be handled in a straightforward manner, usually not requiring any special genetic counseling. If screening is conducted in the more common sequential manner, the majority of women will test negative; and of the few who test positive, the majority of their partners will test negative, and that will be the end of it. If the woman tests positive and her partner also tests positive, prenatal diagnosis by either amniocentesis or chorionic villus sampling will be offered, and

Prenatal Diagnosis of Cystic Fibrosis

623

the molecular test will involve looking for the two parental mutations in the fetus; there is a 1-in-4 chance that both will be found, in which case referral for genetic counseling should be made and the option for pregnancy termination will be offered. However, there are certain situations that require special consideration, such as positive–negative couples, positive family history, echogenic bowel, assisted reproduction and preimplantation diagnosis, and newborn screening. Positive–negative couples

The situation where one member of the couple (usually the mother, if sequential screening is done) tests positive for a CFTR mutation and the other tests negative presents some additional concern. The problem with this scenario is that the couple’s risk to have an affected child is now higher than it was before they were tested, yet the guidelines state that nothing further need be offered to them. The reason is that if the negative partner does carry a mutation, it is a rare one not covered by the ACMG panel (or whatever targeted panel was used) and therefore not detectable in the fetus either. It is not recommended to proceed with targeted prenatal diagnosis in this situation because if the one mutation is found in the fetus, there is no way to know whether the fetus is affected or (more likely) a healthy carrier of just the one mutation. Given the current availability of NGS, however, it may be reasonable to offer a larger panel or even whole-gene sequencing and deletion analysis to the negative partner, in the hopes of identifying (or, conversely, lowering the possibility of) a very rare CFTR mutation. But again, the possibility of identifying a problematic VUS in the other partner always exists when going down this route. Positive family history

Although much of this chapter has been concerned with population-based carrier screening in those with no family history of CF, there are those who will have an affected relative, and it is essential that such family history be sought because it dramatically changes the a priori risks of being a carrier (and in so doing, one should also inquire about a family history of male infertility). Even before the launch of population carrier screening, such testing was always made available to individuals with a positive family history. Moreover, it is perfectly

624 Genetic Disorders and the Fetus

acceptable to use expanded panels in this situation, because one wants to optimize the chances of identifying the familial mutation. Ideally, testing will be done first on the affected index case in the family so that the familial mutation will already be known. The bottom line is that for couples with a positive family history of CF, one should go the extra mile and not rely solely on guidelines for routine population carrier screening. Echogenic bowel

Although we have generally described CF in this chapter as a disease which shows no visible signs in the fetus, that is not entirely true. A small proportion of CF fetuses will show echogenic bowel on ultrasound, though the finding is also seen in a wide variety of other conditions, both acquired (e.g. infection) and congenital, and may be present when there is nothing wrong with the fetus at all.79 Combining the results of several series, it appears that an isolated finding of echogenic bowel carries a 1–2 percent risk that the fetus has CF.80 This is considered high enough to warrant CFTR mutation testing in the fetus. If two mutations are found, the conclusion is straightforward, but if only one is found, there will be much uncertainty and anxiety. This may be one of the few cases where full-gene sequencing and deletion analysis may be warranted in the prenatal screening situation, in order to provide the best possible chance of identifying the second mutation, if it is there.75

“allele dropout” that can produce false results, with disastrous consequences. Another, less expensive option, of course, is to use an unrelated sperm or egg donor (who has already been screened negative for CFTR mutations). Sometimes it will be an adult male CF patient himself who presents for assisted reproductive technologies (ART). The reason, of course, is the problem of CBAVD. Such men are not truly infertile; they do make sperm, but it cannot get out into the ejaculate. However, many of these men have fathered children by sperm aspiration.82 Remember that genetic counseling and partner screening are essential in such cases, because the father has a 100 percent chance of passing a CFTR mutation or less penetrant variant to his offspring. Newborn screening

CF is one of the diseases chosen for expanded newborn screening, and all US states and many countries are now including it, usually by measurement of IRT followed by limited mutation testing. Its many ramifications are beyond the scope of this chapter, except to note that newborn screening is another way in which carrier parents will be identified (indirectly). This knowledge will in turn influence their attitudes toward prenatal diagnosis in a future pregnancy. In this sense, newborn screening and carrier screening represent a dynamic interaction, with each affecting the frequencies and attitudes of the other.83

Assisted reproduction and preimplantation diagnosis

Future directions

As can be gleaned from the discussion in “Outcomes of the CF carrier screening program”, there are many couples who find the prospect of pregnancy termination for CF problematic. For those that do but would still like to avoid having a child with CF, in vitro fertilization followed by preimplantation genetic testing (PGT) by single-cell biopsy of the embryo is a possible option. It is expensive and not always covered by insurance, so it is largely available only to those couples who can afford it. But it is certainly feasible, as long as both parental CFTR mutations are known from previous testing. In fact, CF was the first disease for which PGT was successfully performed.81 The technique is available in only a handful of centers and is technically challenging, subject to artifacts like

In addition to the other strengths of NGS discussed in this chapter, we cannot conclude without at least mentioning the NGS test most familiar to obstetricians: noninvasive prenatal screening (NIPS). Its advantages over traditional prenatal specimen collection by amniocentesis and CVS are well known, allowing for testing earlier in the pregnancy and at no risk to the fetus (see Chapter 8). Its primary application has been in screening for trisomies and other aneuploidies, for which it boasts far better predictive value than maternal serum screening.84 Since it involves whole-genome sequencing, it can also theoretically go beyond merely counting chromosome “reads” to detect subchromosomal deletions and insertions, and some NIPS laboratories now claim to screen for several

CHAPTER 15

known syndromes of that type, such as 22q11 deletion/DiGeorge syndrome. Extending the resolution even farther, it should be possible to sequence and reconstruct the entire fetal genome, opening the way for noninvasive prenatal diagnosis of single-gene disorders such as CF.85 Although some laboratories have begun offering such tests for a variety of disorders, including CF,86 it is not without significant challenges, most prominently the need to tease out a fetal sequence change from the much more abundant background of maternal DNA.87 Nevertheless, given the rapid pace of advances in technology, it is reasonable to predict that such capabilities will improve enough for routine use in the next few years. At that point, for pregnant women already being offered NIPS for aneuploidy detection, CF and any number of other single-gene disorders could be added in at that time, obviating the need for prior carrier screening of the parents. In this way, NIPS could become the universal reproductive screening test, assimilating much of the purpose of both newborn and carrier screening. Further in the future, we can expect a proteomics revolution analogous to the genomics revolution which we are now living through. Its complexity can only be imagined at this point, but

Prenatal Diagnosis of Cystic Fibrosis

625

one of its fruits will hopefully be a more definitive understanding of the effect of gene mutations at the protein level – this would be a boon to both carrier screening and prenatal diagnosis of CF, where our current ability to predict genotype–phenotype correlations is less than optimal. It would also presumably help to identify and characterize the function of CFTR-modifier genes.88 Will CF as a disease ever be completely eliminated through carrier screening and prenatal diagnosis? Such a goal is virtually impossible for any recessively inherited disease. Hypothetically, it would require truly universal uptake of parental carrier screening and 100 percent of at-risk couples choosing to terminate affected pregnancies, something we are unlikely to see with this particular disease. Some ethicists might argue that it is not even the goal we should be striving for. Our mission in this area, as in all aspects of medical genetics, should always be to provide patients with the most current, unbiased information and the full range of interventional options open to them. Thanks to the power of the modern molecular genetic technologies, we can certainly do that in a way that previous generations could hardly have conceived.

References 1. Bobadilla J, Macek M, Fine JP, et al. Cystic fibrosis:

8. Vricella LA, Karamichalis JM, Ahmad S, et al. Lung and

a worldwide analysis of CFTR mutations – correlation

heart-lung transplantation in patients with end-stage

with incidence data and application to screening. Hum

cystic fibrosis: the Stanford experience. Ann Thorac

Mutat 2002;19:575. 2. Farrell PM. The prevalence of cystic fibrosis in the European Union. J Cyst Fibrosis 2008;7:450. 3. Tonelli MR, Aitken ML. Pregnancy and cystic fibrosis. Curr Opin Pulm Med 2007;13:537. 4. Ahmad A, Ahmed A, Patrizio P. Cystic fibrosis and fertility. Curr Opin Obstet Gynecol 2013;25:167. 5. Farrell PM, White TB, Ren CL, et al. Diagnosis of cystic fibrosis: consensus guidelines from the Cystic Fibrosis Foundation. J Pediatr 2017;181S:S4. 6. Gregg RG, Simantel A, Farrell PM, et al. Newborn

Surg 2002;74:13. 9. Maule G, Arosio D, Cereseto A. Gene therapy for cystic fibrosis: progress and challenges of genome editing. Int J Mol Sci 2020;21:E3903. 10. Derichs N. Targeting a genetic defect: cystic fibrosis transmembrane conductance regulator modulators in cystic fibrosis. Eur Respir Rev 2013;22:58. 11. Ramsey BW, Davies J, McElvaney NG, et al. A CFTR potentiator in patients with cystic fibrosis and the G551D mutation. N Engl J Med 2011;365:1663.

screening for cystic fibrosis in Wisconsin: compari-

12. Egan ME. Cystic fibrosis transmembrane conductance

son of biochemical and molecular methods. Pediatrics

receptor modulator therapy in cystic fibrosis, an update.

1997;99:819.

Curr Opin Pediatr 2020;32:384.

7. Bilton D, Stanford G. The expanding armamentarium of

13. Massie J, Castellani C, Grody WW. Carrier screening for

drugs to aid sputum clearance: how should they be used

cystic fibrosis in the new era of medications that restore

to optimize care? Curr Opin Pulm Med 2014;20:601.

CFTR function. Lancet 2014;383:923.

626 Genetic Disorders and the Fetus

14. Riordan JR, Rommens JM, Kerem B, et al. Identification of the cystic fibrosis gene: cloning and characterization of complementary DNA. Science 1989;245:1066. 15. Rommens JM, Iannuzzi MC, Kerem B, et al. Identification of the cystic fibrosis gene: chromosome jumping and walking. Science 1989;245:1059. 16. Hasegawa HW, Skach W, Baker O, et al. A multifunctional aqueous channel formed by CFTR. Science 2008;258:1477. 17. Ogino S, Gulley ML, den Dunnen JT, Wilson RB, and the Association for Molecular Pathology Training and Education Committee. Standard mutation nomenclature in molecular diagnostics: practical and educational challenges. J Mol Diagn 2007;9:1-6 18. Tsui L-C, Dorfman R. The cystic fibrosis gene : a molecular genetic perspective. Cold Spring Harb Perspect Med 2013;3:a009472. 19. Tsui L-C. The spectrum of cystic fibrosis mutations. Trends Genet 1992;8:392. 20. Kerem BS, Buchanan JA, Durie P, et al. DNA marker haplotype association with pancreati sufficiency in cystic fibrosis. Am J Hum Genet 1989;44:827. 21. Kristidis P, Bozon D, Corey M, et al. Genetic determination of exocrine pancreatic function in cystic fibrosis. Am J Hum Genet 1992;50:1178. 22. Kerem E, Corey M, Kerem B, et al. The relation between genotype and phenotype in cystic fibrosis: analysis of the most common mutation (deltaF508). N Engl J Med 1990;323:1517. 23. Cutting GR, Kasch LM, Rosenstein BJ, et al. Two patients with cystic fibrosis, nonsense mutations in each cystic fibrosis gene, and mild pulmonary disease. N Engl J Med 1990;323:1685. 24. Burke W, Aitken JL, Chen S-H, Scott DR. Variable severity of pulmonary disease in adults with identical cystic fibrosis mutations. Chest 1992;102:506. 25. Cystic Fibrosis Genotype-Phenotype Consortium. Correlation between genotype and phenotype in patients with cystic fibrosis. N Engl J Med 1993;329:1308. 26. Highsmith WE, Burch LH, Zhou Z, et al. A novel mutation in the cystic fibrosis gene in patients with pulmonary diseae but normal sweat chloride concentrations. N Engl J Med 1994;331:974-980. 27. Wilschanski M, Zielenski J, Markiewicz D, et al. Correlation of sweat chloride concentration with classes of the cystic fibrosis transmembrane conductance regulator gene mutations. J Pediatr 1995;127:705. 28. Gervais R, Dumur V, Rigot M-M, et al. High frequency of the R117H cystic fibrosis mutation in patients with congenital absence of the vas deferens. N Engl J Med 1993;328:446.

29. Cutting GR. Modifier genetics: cystic fibrosis. Annu Rev Genom Hum Genet 2005;6:237. 30. Anguiano A, Oates RD, Amos J, et al. Congenital bilateral absence of the vas deferens: a primarily genital form of cystic fibrosis. JAMA 1992;267:1794. 31. Mercier B, Verlingue C, Lissens W, et al. Is congenital bilateral absence of vas deferens a primary form of cystic fibrosis? Analyses of the CFTR gene in 67 patients. Am J Hum Genet 1995;56:272. 32. Kerem E, Rave-Harel N, Augarten A, et al. A cystic fibrosis transmembrane conductance regulator splice variant with partial penetrance associated with variable cystic fibrosis presentations. Am J Respir Crit Care Med 1997;155:1914. 33. Kiesewetter S, Macek M Jr,, Davis C, et al. A mutation in the CFTR produces different phenotypes depending on chromosomal background. Nat Genet 1993;5:274. 34. Massie RJH, Poplawski N, Wilcken B, et al. Intron-8 polythymidine sequence in Australasian individuals with CF mutations R117H and R117C. Eur Respir J 2001;17:1195. 35. Lebo RV, Grody WW. Variable penetrance and expressivity of the splice altering 5T sequence in the cystic fibrosis gene. Genet Test 2007;11:32. 36. Groman JD, Hefferon TW, Casals T, et al. Variation in a repeat sequence determines whether a common variant of the cystic fibrosis transmembrane conductance regulator gene is pathogenic or benign. Am J Hum Genet 2004;74:176. 37. Collaco JM, Vanscoy L, Bremer L, et al. Interactions between second-hand smoke and genes that affect cystic fibrosis lung disease. JAMA 2008;299:417. 38. O’Neal WK, Knowles MR. Cystic fibrosis disease modifiers: complex genetics defines the phenotypic diversity in a monogenic disease. Annu Rev Genomics Hum Genet 2018;19:201. 39. Macek M, Mackova A, Hamosh A, et al. Identification of common cystic fibrosis mutations in AfricanAmericans with cystic fibrosis increases the detection rate to 75%. Am J Hum Genet 1997;60:1122. 40. Palomaki GE, FitzSimmons SC, Haddow JE. Clinical sensitivity of prenatal screening for cystic fibrosis via CFTR carrier testing in a United States panethnic population. Genet Med 2004;6:405. 41. Shoshani T, Augarten A, Gazit E, et al. Association of a nonsense mutation (W1282X), the most common mutation in the Ashkenazi Jewish cystic fibrosis patients in Israel, with presentation of severe disease. Am J Hum Genet 1992;50:222. 42. Workshop on Population Screening for the Cystic Fibrosis Gene. Statement from the National Institutes of

CHAPTER 15

43.

44.

45.

46.

47.

48.

49.

50.

51.

52.

53.

54.

55. 56.

Health workshop on population screening for the cystic fibrosis gene. N Engl J Med 1990;323:70. Caskey CT, Kaback MM, Beaudet AL. The American Society of Human Genetics statement on cystic fibrosis screening. Am J Hum Genet 1990;46:393. Tambor ES, Bernhardt BA, Chase GA, et al. Offering cystic fibrosis carrier screening to an HMO population: factors associated with utilization. Am J Hum Genet 1994;55:626. Loader S, Caldwell P, Kozyra A, et al. Cystic fibrosis carrier population screening in the primary care setting. Am J Hum Genet 1996;59:234. Clayton EW, Hannig VL, Pfotenhauer JP, et al. Lack of interest by nonpregnant couples in population-based cystic fibrosis carrier screening. Am J Hum Genet 1996;58:617. Grody WW, Dunkel-Schetter C, Tatsugawa ZH, et al. PCR-based screening for cystic fibrosis carrier mutations in an ethnically diverse pregnant population. Am J Hum Genet 1997;60:935. NIH Consensus Statement. Genetic Testing for Cystic Fibrosis. NIH Consensus Statement Online, April 14–16, 1997. http://consensus.nih.gov/1997/ 1997GeneticTestCysticFibrosis106html.htm Grody WW, Desnick RJ. Cystic fibrosis population carrier screening: here at last – are we ready? Genet Med 2001;3:87. Grody WW, Cutting GR, Klinger KW, et al. Laboratory standards and guidelines for population-based cystic fibrosis carrier screening. Genet Med 2001;3:149. Watson MS, Cutting GR, Desnick RJ, et al. Cystic fibrosis population carrier screening: 2004 revision of American College of Medical Genetics mutation panel. Genet Med 2004;6:387. Rohlfs EM, Zhou Z, Sugarman EA, et al. The I148T CFTR allele occurs on multiple haplotypes: a complex allele is associated with cystic fibrosis. Genet Med 2002;4:319. Buller A, Olson S, Redman JB, et al. Frequency of the cystic fibrosis 3199del6 mutation in individuals heterozygous for I148T. Genet Med 2004:6:108. Monaghan KG, Highsmith WE, Amos J, et al. Genotype-phenotype correlation and frequency of the 3199del6 cystic fibrosis mutation among I148T carriers: Results from a collaborative study. Genet Med 2004;6:421. Wald NJ. Couple screening for cystic fibrosis. Lancet 1991;338:1318. Doherty RA, Palomaki GE, Kloza EM, et al. Couple-based prenatal screening for cystic fibrosis in primary care settings. Prenat Diagn 1996;16:397.

Prenatal Diagnosis of Cystic Fibrosis

627

57. Super M, Schwartz MJ, Malone G, et al. Active cascade testing for carriers of cystic fibrosis gene. Lancet 1991;338:1318. 58. Castellani C, Duff B, Dalla Piccola, B, et al. Benchmarks for cystic fibrosis carrier screening: a European consensus document. J Cyst Fibrosis 2010;9:165. 59. Morgan MA, Driscoll DA, Mennuti MT et al. Practice patterns of obstetrician-gynecologists regarding preconception and prenatal screening for cystic fibrosis. Genet Med 2004;6:450. 60. Morgan MA, Driscoll DA, Zinberg S, et al. Impact of self-reported familiarity with guidelines for cystic fibrosis carrier screening. Obstet Gynecol 2005;105:1355. 61. Brennan ML, Schrijver I. Cystic fibrosis: a review of associated phenotypes, use of molecular diagnostic approaches, genetic characteristics, progress, and dilemmas. J Mol Diagn 2016;18:3. 62. Palomaki GE, Bradley LA, Richards CS, Haddow JE. Analytic validity of cystic fibrosis testing: a preliminary estimate. Genet Med 2003;5:15. 63. Lyon E, Schrijver I, Weck KE, et al. Molecular genetic testing for cystic fibrosis: laboratory performance on the College of American Pathologists external proficiency surveys. Genet Med 2015;17:219. 64. Heim RA, Sugarman EA, Allitto BA. Improved detection of cystic fibrosis mutations in the heterogeneous U.S. population using an expanded, pan-ethnic mutation panel. Genet Med 2001;3:168. 65. Amos JA, Bridge-Cook P, Ponek V, Jarvis MR. A universal array-based multiplexed test for cystic fibrosis carrier screening. Expert Rev Mol Diagn 2006;6:15. 66. Alper OM, Wong LJ, Young S, et al. Identification of novel and rare mutations in California Hispanic and African American cystic fibrosis patients. Hum Mutat 2004;24:353. 67. Schrijver I, Ramalingam S, Sankaran R, et al. Diagnostic testing by CFTR gen mutation analysis in a large group of Hispanics: Novel mutations and assessment of a population-specific mutation spectrum. J Mol Diagn 2005;7:289. 68. Claustres M, Altiéri JP, Guittard C, et al. Are p.I148T, p.R74W and p.D1270N cystic fibrosis causing mutations? BMC Med Genet 2004;5:19. 69. Grody WW. Expanded carrier screening and the law of unintended consequences: from cystic fibrosis to fragile X. Genet Med 2011;13:996. 70. Strom CM, Redman JB, Peng M. The dangers of including nonclassical cystic fibrosis variants in population-based screening panels: p.L997F, further genotype/phenotype correlation data. Genet Med 2011;13:1042.

628 Genetic Disorders and the Fetus

71. Grody WW, Cutting GR, Watson MS. The cystic fibrosis mutation “arms race”: When less is more. Genet Med 2007;9:739. 72. Grosu DS, Hague L, Chelliserry M, et al. Clinical investigational studies for validation of a next-generation sequencing in vitro diagnostic device for cystic fibrosis testing. Expert Rev Mol Diagn 2014;14:605. 73. Sosnay PR, Siklosi KR, Van Goor F, et al. Defining the disease liability of variants in the cystic fibrosis transmembrane conductance regulator gene. Nat Genet 2013;45:1160. 74. Richards S, Aziz N, Bale S, et al.; ACMG Laboratory Quality Assurance Committee. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med 2015;17:405. 75. Deignan JL, Astbury C, Cutting GR, et al.; ACMG Laboratory Quality Assurance Committee. CFTR variant testing: a technical standard of the American College of Medical Genetics and Genomics (ACMG). Genet Med 2020;22:1288. 76. Grody WW. Where to draw the boundaries for prenatal carrier screening. JAMA 2016;316:717. 77. Witt DR, Schaefer C, Hallam P, et al. Cystic fibrosis heterozygote screening in 5,161 pregnant women. Am J Hum Genet 1996;58:823. 78. Kessels SJM, Carter D, Ellery B, et al. Prenatal genetic testing for cystic fibrosis: a systematic review of clinical effectiveness and an ethics review. Genet Med 2020;22:258. 79. Saha E, Mullins EW, Paramasivam G, et al. Perinatal outcomes of fetal echogenic bowel. Prenat Diagn 2012;32:758.

80. Carcopino X, Chaumoitre K, Shojai R, et al. Fetal magnetic resonance imaging and echogenic bowel. Prenat Diagn 2007;27:272. 81. Handyside AH, Lesko JG, Tarin JJ, et al. Birth of a normal girl after in vitro fertilization and preimplantation diagnostic testing for cystic fibrosis. N Engl J Med 1992;327:905. 82. Josserand RN, Bey-Omar F, Rollet J, et al. Cystic fibrosis phenotype evaluation and paternity outcome in 50 males with congenital bilateral absence of vas deferens. Hum Reprod 2003;16:2093. 83. Hale JE, Parad RB, Comeau AM. Newborn screening showing decreasing incidence of cystic fibrosis. N Engl J Med 2008;358:973. 84. Ashwood ER, Palomaki GE. A new era in noninvasive prenatal testing. N Engl J Med 2013;369:2164. 85. Lo YM. Noninvasive fetal whole-genome sequencing from maternal plasma: feasibility studies and future directions. Clin Chem 2013;59:601. 86. Chandler NJ, Ahlfors H, Drury S, et al. Noninvasive prenatal diagnosis for cystic fibrosis: Implementation, uptake, outcome, and implications. Clin Chem 2020;66:207. 87. Tsao DS, Silas S, Landry BP, et al. A novel high-throughput molecular counting method with single base-pair resolution enables accurate single-gene NIPT. Sci Rep 2019;9:14382. 88. Lim SH, Legere EA, Snider J, Stagljar I. Recent progress in CFTR interactome mapping and its importance for cystic fibrosis. Front Pharmacol 2018;8:997.

16

Prenatal Diagnosis and the Spectrum of Involvement from Fragile X Mutations Randi J. Hagerman and Paul J. Hagerman UC Davis Health System, Sacramento, CA, USA

Introduction Mutations of the fragile X mental retardation 1 (FMR1) gene, including both premutation (55–200 repeats) and full-mutation (>200 repeats) CGG-repeat expansions, give rise to a broad spectrum of cognitive impairment, which ranges from intellectual disability and autism to mild learning or emotional difficulties in the context of normal IQ. In addition, developmental and late adult-onset neurological, cognitive, psychiatric, and medical problems arise in some premutation carriers (fragile X-associated tremor ataxia syndrome (FXTAS)).1–3 Clinical involvement in individuals with the full mutation (fragile X syndrome (FXS)) is a consequence of transcriptional silencing of the gene and the resulting deficiency or absence of the FMR1 protein (FMRP), an RNA-binding protein that transports and regulates the translation of many messages into their respective proteins. The absence of FMRP leads, in turn, to dysregulation of a number of proteins important for synaptic maturation and plasticity.4 Since FMRP specifically downregulates the translation of a number of postsynaptic proteins, production of these proteins is significantly upregulated in the absence of FMRP.5 One important pathway upregulated in the absence of FMRP is the metabotropic glutamate receptor 5 (mGluR5) system, resulting in

long-term depression (LTD) of synaptic activity and weakening of synaptic connections.4, 6 The associated neuroanatomical phenotype includes long, thin (“immature”) synaptic connections, which are thought to be the cause of the intellectual disability in FXS. Recent research has led to a number of treatment trials for FXS using various mGluR5 antagonists, GABA agonists, metformin, and cannabidiol (CBD) which have been shown to reverse at least some of the neuroanatomical and clinical phenotype of FXS in animal models.6, 7 For CGG-repeat expansions in the premutation range, both clinical involvement and its pathogenesis (elevated FMR1 mRNA) are distinct from the FMRP-deficit model for full-mutation alleles and FXS. The molecular pathogenesis for premutation-associated clinical involvement involves a toxic gain-of-function of the expanded-repeat FMR1 mRNA,3, 8 which is produced at elevated levels in the premutation range, in contrast to the reduced/absent levels of FMR1 mRNA in the full-mutation range. However, some patients who are in the upper premutation range or in the full-mutation range without methylation can have both lowered FMRP and elevated FMR1 mRNA, also termed a double hit or a dual mechanism of involvement.3 Although most individuals with the premutation possess normal intellectual abilities, some individuals do experience developmental problems

Genetic Disorders and the Fetus: Diagnosis, Prevention, and Treatment, Eighth Edition. Edited by Aubrey Milunsky and Jeff M. Milunsky. © 2021 Aubrey Milunsky and Jeff M. Milunsky. Published 2021 by John Wiley & Sons Ltd.

629

630 Genetic Disorders and the Fetus

that include attention deficit hyperactivity disorder (ADHD) and/or social deficits, ranging from social anxiety to autism spectrum disorder (ASD).9–12 These problems are more common in males (both children and adults) than in females, although the psychiatric problems, including anxiety and depression, are common in women and the term fragile X-associated neuropsychiatric disorder (FXAND) is an umbrella term to encompass these neuropsychiatric disorders.13 In addition, some adults with the premutation may develop clinical symptoms related to FXTAS,14, 15 including neuropathy;16, 17 autoimmune problems, such as fibromyalgia and hypothyroidism;18 emotional difficulties, including depression and anxiety;19, 20 and dementia.21, 22 The abnormally elevated mRNA triggers a cascade of events in neural cells that ultimately leads to the clinical involvement described later in this chapter.8, 23 The complexity of clinical involvement and treatment, including the emerging targeted treatments that are becoming available for fragile X-related disorders, complicates the genetic counseling aspects of these disorders. This chapter will review epidemiology, clinical involvement, genetic counseling, prenatal diagnostic procedures, and treatment in the fragile X spectrum disorders.

Epidemiology Newborn screening studies in the United States and internationally have found a high prevalence of the premutation in the general population: 1 in 156–250 females and 1 in 250–810 males, with variability depending on the location of the screening (reviewed in refs).24–26 A 2014 review of prevalence studies places the prevalence of the full mutation at approximately 1 in 5,000 to 7,000;27 however, newborn screening studies have not yet confirmed this prevalence because of the large numbers needing to be screened.24 Prevalence may vary in different parts of the world where pockets of families with FXS, presumably from a founder effect, are found; this situation is more common in developing countries such as Ricaurte, Colombia, and Samarang, Indonesia, where poverty may constrain people to the region where they were born. One important correlate with the frequency estimate for male carriers is that as many as ∼1/3,000 males over 50

years in the general population may suffer from FXTAS, since about 40 percent of male carriers may be affected by FXTAS. Also, if we define FXS or the fragile X phenotype as the presence of lowered FMRP leading to developmental problems, then a subgroup of premutation carriers on the high end of the CGG spectrum may be labeled as FXS, even though they do not have a full mutation.28 The emergence of data demonstrating that some individuals with neurodevelopmental or neuropsychiatric problems have lowered FMRP levels in blood and/or brain but without a fragile X mutation has stimulated research regarding FMRP levels across neurodevelopmental disorders.6 Examples of such disorders include schizophrenia, where the age of onset and IQ both correlate with a level of FMRP that is lower than that seen in the general population.29, 30 In addition, Fatemi and colleagues have found lowered FMRP levels in the brain of people with neuropsychiatric conditions, including bipolar disorder, depression, autism, and schizophrenia.31–33 The FMRP function may also be dysregulated by hypoxia and seizures.34 Following early life seizures in the rat, FMRP moves away from the dendrites and localizes near the nucleus so that synaptic plasticity is dysregulated without FMRP at the synapse.34 The increase in seizure frequency can worsen the severity of autism from any cause, including tuberous sclerosis, neurofibromatosis, FXS, premutation involvement, and even idiopathic autism.35, 36 As FMRP regulates the translation of approximately 30 percent of all of the genes associated with autism, particularly those involved with synaptic plasticity,37 the dysregulation of FMRP by seizures would possibly dysregulate many other genes associated with autism, and therefore the social deficits would increase.36 Further populations likely will be found with a deficit of FMRP that go beyond those with an FMR1 mutation, so our definition of what constitutes the fragile X phenotype may be broadened substantially in the future.

Clinical involvement in those with the full mutation Most males with FXS have intellectual disability with a mean IQ in the 40s,38 though the floor effect of most IQ assessment measures does not accurately score IQs below 45.39 Only approximately 15

CHAPTER 16

Prenatal Diagnosis and the Spectrum of Involvement from Fragile X Mutations

percent of males with FXS will have an IQ above 70; these individuals have either significant mosaicism (high percentage of cells with the premutation in addition to the full mutation) or a lack of methylation of the full-mutation alleles.40, 41 Differences in methylation status are often found between blood leukocytes and fibroblasts, but the level of FMRP in either tissue correlates positively with the IQ, and the level of methylation correlates inversely with the level of FMRP.42 Prenatal diagnostic techniques cannot determine the methylation status of the fetus because methylation may not set in for the full mutation until later in gestation, after prenatal testing is carried out. Both the European EMQN guidelines for fragile X testing43 and the American College of Medical Genetics Standards and Guidelines for fragile X testing26 recommend avoiding methylation testing in prenatal diagnosis, particularly chorionic villus sampling (CVS). If an expanded allele is detected, then amniocentesis is recommended to confirm either a premutation or a full-mutation allele.26 It is therefore impossible to determine the level of cognitive involvement in a fetus with the full mutation beyond knowing the range of scores in the males and females with FXS.26 Approximately 70 percent of females with the full mutation will have an IQ of 85 or lower.44 Although only 25 percent of girls with the full mutation will have an IQ lower than 70, those with a borderline IQ (70–85) have significant learning problems, including executive function deficits, ADHD, language delays, impulsivity, visual–spatial perceptual deficits, and academic delays, particularly in math.38, 45–49 These individuals typically require significant interventions during their schooling.50 Approximately 25 percent of females with the full mutation will have a normal IQ without learning disabilities, though emotional problems, such as anxiety, are still common in this group.51 Recent quantitative methods to measure FMRP include western blot,52, 53 enzyme-linked immunosorbent assay (ELISA),54 fluorescence resonance energy transfer (FRET),28, 55 and a Luminex assay.56 The last three techniques are more quantitative than western blot analysis or earlier immunocytochemical methods and are best used to detect mild deficits of FMRP in carriers or in other disorders without an FMR1 mutation.29 As these techniques move into clinical use, those at risk for FMRP deficits will be more easily

631

recognized. Further research will be stimulated with regard to the medications or environmental changes that can upregulate FMRP levels. Psychopathology is common in those with the full mutation and includes ADHD in approximately 80 percent of boys and 25 percent of girls;57, 58 autism or ASD in 60 percent of boys and 10–20 percent of girls;59–63 anxiety or mood disorder in 25–70 percent of boys and girls;64 and psychosis in less than 10 percent.57 Often the behavioral problems, particularly impulsivity, anxiety, mood instability, and aggression, cause the main problems for the family caring for children with FXS. Maternal stress is associated with more severe behavioral problems and anxiety in children with FXS, and mothers with the lowest activation ratio are at greatest risk for stress.65 These difficulties in children with the full mutation often lead to medical interventions and diagnosis, which occur typically between 2.5 and 4 years of age.66, 67 There are many medications and behavioral interventions that can help with these behavioral problems,68 and new targeted treatments for FXS have shown some promising results;6, 7, 69 however, a multidisciplinary intervention plan is necessary, including special education supports and therapies, such as speech and language therapy, occupational therapy, and psychotherapy.70, 71 The physical features of FXS classically include a long face, prominent ears, hyperextensible finger joints, flat feet, and macro-orchidism at puberty. Approximately 30 percent of young children do not have these features, so the diagnosis is often based on behavioral features, such as poor eye contact, hand flapping, hand biting, perseverative speech, autistic features, anxiety, and ADHD symptoms.72 Many individuals are diagnosed with autism or ASD before the diagnosis of FXS, so all children with ASD should have FMR1 testing.72 The medical problems associated with FXS are relatively few, and most are caused by the connective-tissue problem that is intrinsic to FXS. The hyperextensible joints on occasion lead to dislocation, but this occurs in fewer than 5 percent of individuals. Hernias are more common (15 percent); in males, the weight of the large testicles combined with the loose connective tissue lead to the hernias. Recurrent otitis media is the most common medical problem (85 percent), followed by strabismus (36 percent), and seizures (20

632 Genetic Disorders and the Fetus

percent).57, 73 The lack of FMRP leads to a seizure phenotype in the fragile X knockout (KO) mouse that improves with the use of mGlur5 antagonists or other targeted treatment for FXS, as described below.74

Clinical phenotype in the premutation The premutation was originally thought to have no phenotype, and males with the premutation were previously described as nonpenetrant males or normal transmitting males (NTMs). Then in 1991, premature ovarian failure (POF; menses stopping before age 40) was described in approximately 20 percent of premutation daughters of these males.75 Subsequent studies have found that this problem increases with increasing CGG-repeat numbers in the premutation carriers, but the prevalence of POF decreases somewhat after 120 repeats.76 This problem has been renamed fragile X-associated primary ovarian insufficiency (FXPOI) because some women with this diagnosis may subsequently become pregnant. The cause of FXPOI seems to be related to the toxicity of elevated FMR1 mRNA on the ovum or the supporting cells of the ovum. For a number of years, psychological problems in male and female carriers with the premutation have been reported with some controversy.77–79 With the identification of FXTAS, there was a clear neurological phenotype, involving an intention tremor, ataxia, neuropathy, autonomic dysfunction, and cognitive decline in some older males and occasional females with the premutation.80 The incidence of FXTAS increases with increasing age, such that only approximately 15 percent of males in their 50s have symptoms, but 75 percent of males in their 80s develop FXTAS.81 In females, approximately 16 percent of carriers older than 50 years develop FXTAS but their symptoms are less severe and cognitive decline is rare.3, 18, 82 Brain atrophy combined with white matter disease is part of the diagnostic criteria in FXTAS,83 with approximately 60 percent of males demonstrating a characteristic sign of increased T2 signal intensity in the middle cerebellar peduncles (MCP sign). The MCP sign is seen, however, in only 13 percent of females with FXTAS.84 Further radiological studies have demonstrated white matter disease in the splenium of the corpus callosum in approximately 50 percent

of patients with FXTAS, so this finding has been added to the diagnostic criteria of FXTAS.85, 86 Additional reports have expanded the phenotype in women with the premutation to include hypothyroidism (50 percent) and fibromyalgia (40 percent) in those with neurological symptoms, suggesting that an autoimmune component occurs in some female carriers.18, 87 For example, multiple sclerosis (MS) is found in approximately 2–3 percent of carriers.18 MS was reported in addition to FXTAS in one case documented at autopsy.86 The elevated mRNA in the premutation leads to upregulation of a number of proteins including 𝛼B-crystallin, which is a primary antigen for MS.88 There is increasing evidence that neurodevelopmental problems are associated with a subgroup of children, particularly boys, with the premutation. Reports of ADHD, social anxiety, and ASD are common in boys with the premutation who present as the proband of the family,9, 10, 67, 89, 90 and although most have a normal IQ, some have intellectual disability.91 The presence of seizures in premutation boys is associated with both ASD and intellectual disability.15 In carriers with ASD, intellectual disability, and/or neurological problems, approximately 20 percent demonstrate a second genetic hit, which is likely to have an additional and cumulative deleterious effect on the individual.92 Premutation neuronal cell cultures demonstrate decreased branching, fewer synaptic connections, earlier cell death, and slower moving mitochondria compared with control neurons.93, 94 In the retina, FXS mice show a 47 percent deficit of rhodopsin and deficits in retinal stimulation,95 which may relate to the visual–perceptual deficits that have been demonstrated in babies and toddlers with the full mutation and also the premutation.96

Pathogenesis of the premutation-associated disorder FXTAS Of the premutation-associated disorders, we understand the most about the pathogenic mechanisms linked to FXTAS; however, similar mechanisms may be at play for both the neurodevelopmental and neuropsychiatric involvement covered under FXAND and in the early menopausal features (FXPOI).13, 97, 98 Therefore, the following paragraphs will focus on FXTAS.

CHAPTER 16

Prenatal Diagnosis and the Spectrum of Involvement from Fragile X Mutations

Neuropathology

Gross neuropathological features of the brains examined postmortem from individuals who had died with FXTAS reveal a general loss of brain volume, with prominent white matter disease involving pallor and spongioform changes, accompanied by a loss of axons and myelin. The regions of pallor are associated with regions of high signal on T2-weighted magnetic resonance images (MRIs) in the same individuals.84, 99–101 The primary neuropathological finding is the presence of solitary, spherical (1–5 μm) ubiquitin-positive intranuclear inclusions in both neurons and astrocytes in broad distribution throughout the brain.100, 101 The greatest concentration of inclusions is found in the hippocampal formation (up to 40 percent of nuclei-bearing inclusions in some cases), with lower inclusion densities (2–10 percent) in cortical neurons and the near absence of inclusions in Purkinje cells of the cerebellum, despite substantial Purkinje cell dropout. Inclusion counts are highly correlated with the number of CGG repeats within the premutation range.101 More recently, inclusions have also been observed in tissues outside of the central nervous system, including in both anterior and posterior pituitary, in the Leydig and myotubular cells of the testes,102 and in ganglion cells of adrenal medulla, dorsal root ganglia, paraspinal sympathetic ganglia, mesenteric ganglia, and subepicardial autonomic ganglia.103 Finally, the presence of inclusions in neuronal nuclei within the hypoglossal cranial nerve nucleus may represent a neuropathological correlate to the late-stage swallowing difficulties experienced by many FXTAS patients.101 Molecular pathogenesis

Several lines of evidence indicate that the pathogenesis of FXTAS involves a “toxic” gain-of-function of the FMR1 mRNA. First, FXTAS is almost exclusively confined to carriers of premutation alleles, where the gene is active; although FXTAS has been reported in carriers with rare, unmethylated full mutations, where the gene is still active.104–106 This observation indicates that FXTAS is not the result of the loss of FMRP, because FMRP levels are only moderately lowered within the premutation range, yet profoundly lowered or absent in individuals with full mutation, fully methylated FMR1 alleles (i.e. those with FXS). Second, the absence of FXTAS

633

among individuals with large, full-mutation, fully methylated alleles (e.g. >500–1,000 repeats) indicates that the CGG-repeat expansion, as DNA, is not contributing to disease formation. Third, the FMR1 mRNA is abnormal in at least two important respects, namely, that its production is substantially increased in the premutation range,52, 107, 108 and that it possesses the expanded CGG-repeat element. Additionally, several important features of the disorder (neurodegenerative changes and the presence of inclusions) have been recapitulated in both mouse and Drosophila models of FXTAS.109–111 Although the mechanism(s) by which the expanded-repeat mRNA triggers the molecular pathogenesis of FXTAS has not been established, at least two models have been put forward (Figure 16.1). In the first model, the expanded CGG-repeat element as RNA binds increased numbers of specific proteins, thereby “sequestering” those proteins from their normal cellular roles. A precedent for this model is the sequestration of muscleblind-like protein 1 (MBNL1) by the expanded CUG-repeat element in the 3′ untranslated region of the myotonic dystrophy protein kinase (DMPK) gene of myotonic dystrophy.112–114 For FXTAS, a number of candidate proteins have been identified and listed in table 1 of reference 115 .116–120 The most candidates for sequestration are the pair of proteins (DGCR8 and DROSHA) comprising the “microprocessor” that processes micro (mi)RNA precursors in the nucleus.121 Diminished levels of a broad range of miRNAs, and increased levels of their precursors, are observed in the brains of FXTAS cases, and restoration of DGCR8 and DROSHA in cell culture models is capable of reversing some of the cellular defects. However, a specific link between specific miRNAs and FXTAS has not been established. A second model for FXTAS pathogenesis is the aberrant translation of the expanded CGG-repeat region from a non-AUG start codon upstream of the CGG-repeat element. This process, termed repeat-associated non-AUG (RAN) translation, generates a peptide, FMRpolyG, that contains a polyglycine element that is translated from the +1 (GGC) codon frame.122–124 In normal translation of FMRP, a canonical AUG start codon downstream of the CGG repeat is used; thus, the CGG (+0 frame) is not translated. Although FMRpolyG

634 Genetic Disorders and the Fetus FMR1 gene

CGGn

AUG

FMRP coding

5′ UTR

3′ UTR

15 mm) (Figure 17.7). It may be unilateral or bilateral as well as asymmetrical. Hydrocephaly is commonly used when severe ventriculomegaly is present, leading to progressive macrocephaly due

652 Genetic Disorders and the Fetus

(a)

(b)

(c)

(d)

(e) Figure 17.4 (a) Transverse section of the fetal head at the level of the septum cavum pellucidum, demonstrating the “lemon” sign (scalloping of the frontal bones). (b) Suboccipital bregmatic view, demonstrating the “banana” sign (anterior curvature of the cerebellar

hemispheres and obliteration of the cisterna magna). (c–e) Second-trimester fetus at 20 weeks of gestation with open spina bifida visible on coronal plane (c, d) and sagittal plane (e).

CHAPTER 17

Prenatal Diagnosis of Fetal Malformations by Ultrasound 653

(a)

(b)

(c)

(d)

Figure 17.5 A fetus with spina bifida in the first trimester at 13 weeks of gestation. (a) Transverse section of the fetal head with dried up brain. (b) The “crash” sign. (c) Sagittal plane showing the absence of intracranial translucency. (d) Mid-sagittal plane presenting the spina bifida.

to increased intracranial pressure, usually caused by obstruction. The level of the obstruction is defined by examining the aqueduct of Sylvius and the third and fourth ventricles. When spina bifida is present, macrocephaly may be absent, since in those cases the head can be relatively small. Ventriculomegaly is a relatively frequent finding during the 20 week scan with a prevalence of 1:675.40 The prevalence of congenital hydrocephalus is more difficult to define, mostly because the clinical criteria and definition vary between studies. The prevalence has been reported as between 1 and 32 per 10,000 live births.41 When a prenatal diagnosis of fetal ventriculomegaly is made prenatal counseling is challenging since mild ventriculomegaly may not always be an anomaly but rather a sonographic sign. Males are more frequently affected than females.42 Ventriculomegaly may result from a heterogeneous

spectrum of conditions including spina bifida, chromosome anomalies (most frequently Down syndrome),43–45 genetic syndromes,46 intrauterine hemorrhage, and congenital infection such as cytomegalovirus and toxoplasmosis,42 although the majority of cases remain unresolved. In up to 30–50 percent of patients presenting to a fetal medicine unit with ventriculomegaly, additional findings will be present.42, 47 The overall prevalence of abnormal chromosomes is around 5 percent. However, when considering fetuses with and without associated findings separately, the prevalence of chromosome anomalies was 1 in 12 for associated and 1 in 33 for isolated cases.47 Thus, the fetal and neonatal mortality and morbidity and neurological development of ventriculomegaly are strongly related to the presence of other malformations and chromosomal defects. Isolated mild ventriculomegaly is associated with a good prognosis and

654 Genetic Disorders and the Fetus

Figure 17.6 Meckel–Gruber syndrome with the association of multicystic kidneys in a coronal view (left), posterior encephalocele in coronal, sagittal, and axial lanes (middle). and hexadactyly (right).

Source: Ville YG, Bault J-P. Diagnosis of fetal malformations by ultrasound. In: Milunsky A, Milunsky JM, eds. Genetic disorders and the fetus, 7th edn. Hoboken, NJ: John Wiley & Sons, 2016, chapter 13.

significant hydrocephalus is poor, with high fetal wastage or perinatal death due mainly to the associated anomalies. Severe intellectual disability is common among the survivors. Hydranencephaly

Figure 17.7 A fetus with severe ventriculomegaly – hydrocephaly.

the risk of abnormal neurodevelopmental delay is around 8 percent.47 One of the causes of hydrocephalus may be congenital stenosis of the aqueduct of Sylvius. One of the most frequent heritable causes of isolated hydrocephalus in males is a mutation in L1 cell adhesion molecule gene (L1CAM).48 Associated findings are adducted thumbs, although these may be difficult to demonstrate prenatally. Other genetic syndromes associated with ventriculomegaly are the RASopathies (see Chapter 14) (including Noonan and Costello syndrome), ciliopathies (including Joubert and Meckel–Gruber syndrome), megalencephaly-associated syndromes, dystroglycanopathies (Walker–Warburg syndrome), and syndromes associated with skeletal anomalies such as craniosynostosis (Apert) or dwarfism (achondroplasia).46, 49 The prognosis in cases of

Congenital absence of the cerebral hemispheres with preservation of the midbrain and cerebellum may result from widespread vascular occlusion in the distribution of the internal carotid arteries, prolonged severe hydrocephalus, an overwhelming infection such as toxoplasmosis or cytomegalovirus, or defects in embryogenesis.50 The condition is generally not inherited and is usually incompatible with survival beyond early infancy. Ultrasonographically, the complete absence of echoes–resonance from the anterior and middle fossae distinguishes hydranencephaly from severe hydrocephalus, in which a thin rim of remaining cortex and the midline echo can always be identified. Holoprosencephaly

Holoprosencephaly is classically defined as incomplete cleavage of the forebrain (prosencephalon) that originates from failed midline delineation during early development. There are three classic types, from the most to least severe: alobar, semilobar, and lobar and a milder subtype – the middle interhemispheric (MIH) variant.51 Holoprosencephaly is associated with facial anomalies of varying severity ranging from cyclopia and proboscis to ocular hypotelorism. Facial midline defects are also often

CHAPTER 17

Prenatal Diagnosis of Fetal Malformations by Ultrasound 655

Figure 17.8 A fetus with holoprosencephaly at 14.6 weeks of gestation.

present. The prevalence of this brain abnormality is around 1.3 in 10,000 births.7, 52 Holoprosencephaly may be associated with chromosome anomalies in 22 percent of cases of which 14 percent occurred de novo and 8 percent were inherited and caused by parental translocations.53 The most common chromosomal abnormality is trisomy 13. Additionally, another frequent cause of holoprosencephaly are monogenic disorders. The Sonic Hedgehog (SHH) gene was the first discovered holoprosencephaly gene and subsequently many more genes have been involved, all of which encode for proteins belonging to brain development pathways and involved in the regulation of SHH activity.54–56 Prenatal diagnosis by ultrasonography is based on the demonstration of a single dilated midline ventricle replacing the two lateral ventricles or partial segmentation of the ventricles in the standard transverse view of the fetal head for measurement of the biparietal diameter (Figure 17.8). Microcephaly

The definition of microcephaly is usually defined as a head circumference of more than 3 standard deviations (SD) below the age- and sex-related mean. It has a birth incidence of approximately 1 per 10,000 and is commonly found in the presence of other brain abnormalities, such as encephalocele or holoprosencephaly. Prenatal diagnosis is based on the demonstration of a decrease in the head-to-abdomen circumference ratio in the presence of associated abnormal intracranial pathology and normal growth in the abdominal circumference. Another common

feature is a sloping forehead. Since the decrease in head circumference may not become apparent before the third trimester, prenatal diagnosis in the second trimester is low and around 10 percent.30 Microcephaly has been associated with chromosomal defects (trisomy 13 and 18), microdeletions, monogenic disorders, or malformation syndromes. Other etiologic factors include fetal hypoxia, intrauterine infection (such as cytomegalovirus and Zika virus),57, 58 and exposure to radiation or other teratogens, including the anticoagulant warfarin. Autosomal recessive primary microcephaly (MCPH) is a static developmental anomaly and to date multiple MCPH genes have been identified. The inactivation of most MCPH genes leads to neurogenesis defects, while the upregulation of some MCPH genes is associated with different kinds of carcinogenesis.59 Prognosis depends on the underlying cause, but in approximately 90 percent of the cases, there is severe intellectual disability. Corpus callosum agenesis

The corpus callosum is a large interhemispheric tract connecting the cerebral hemispheres and appearing as a hypoechoic structure located between the cavum septum pellucidum and the cingulate gyrus. Both 2D and 3D techniques allow imaging of the corpus callosum from 18–20 weeks of gestation. Prior to 18 weeks the pericallosal arteries can be visualized using color Doppler and this visualization is an indirect sign that a normal corpus callosum will develop.60 Corpus callosum agenesis can be either complete or partial. In complete corpus callosum agenesis both the corpus callosum and the cavum septum pellucidum are absent. The normal pericallosal arteries cannot be identified when using color Doppler. In the axial plane the frontal horns appear narrow and laterally displaced, and the atria and occipital horns are slightly dilated (colpocephaly); the shape is similar to a teardrop.61 On coronal section, the falx cerebri can be seen in a broad interhemispheric fissure which meets the third ventricle; the lateral ventricles are widely separated and vertically oriented (“Viking’s helmet” sign) (Figure 17.9). The suspicion of a corpus callosum agenesis should rise when the cavum septum pellucidum cannot be visualized during the 20 week scan. The cavum septum pellucidum should be

656 Genetic Disorders and the Fetus

(a)

(b)

Figure 17.9 (a) Transverse and (b) coronal plane of corpus callosum agenesis at 21.2 weeks of gestation.

seen in all fetuses at 18 and 37 weeks of gestation. This is specifically a second-trimester diagnosis which can reach detection rates of 95 percent.30 When in doubt, fetal magnetic resonance imaging (MRI) (see Chapter 19) may provide additional information, however, the actual diagnostic accuracy of fetal MRI in isolated corpus callosum agenesis is still debated.62 Corpus callosum agenesis can be an isolated finding or associated with other structural anomalies. In the case of isolated or partial corpus callosum the rate of chromosome anomalies is 5.7 percent.63 The risk of associated anomalies detected at fetal MRI is about 8 percent and 12 percent, whereas associated anomalies detected postnatally can occur in about 5 percent and 14 percent of fetuses with complete and partial corpus callosum agenesis, respectively. Neurodevelopmental outcome in complete corpus callosum agenesis is normal in 76 percent and in partial corpus callosum agenesis in 71 percent.63

Mega cisterna magna

In mega cisterna magna the cisterna magna is >10 mm in an oblique transverse plane which visualizes the cavum septum pellucidum, cerebellum, cisterna magna, and nuchal fold. The cerebellum is normally developed. Even at 11–13 weeks an abnormal appearance of the fourth ventricle–cisterna magna complex is present.65 In a recent systematic review, in 7 percent of cases the prenatal diagnosis of mega cisterna magna was not confirmed postnatally.66, 67 When associated with other structural anomalies, the risk of chromosome anomalies and genetic syndromes is high, with trisomy 18 being the most common.68 Mega cisterna magna was not significantly associated with additional anomalies detected at prenatal MRI or detected after birth. The outcome in isolated cases is generally good.67

Blake’s pouch cyst Posterior fossa malformations

During a 20 weeks anomaly scan the transcerebellar plane allows evaluation of the posterior fossa or the cisterna magna.8 The size of the cisterna magna is around 5 mm with an upper limit of 10 mm.64 An increased amount of fluid in the cisterna magna should raise the suspicion of pathology. Posterior fossa malformations embody a heterogeneous spectrum of conditions. It is difficult to delineate the prenatal detection rate in these malformations, since isolated cases may have a completely normal outcome and may remain undiagnosed during life.

The Blake’s pouch is an embryological cystic structure bulging into the fourth ventricle (Figure 17.10). When fenestration fails or is delayed, a cyst may develop. Resolution of the cyst may occur until 26 weeks. The prenatal diagnosis is false positive in up to 10 percent of cases. The rates of associated central nervous system and other structural anomalies are 12 percent and 25 percent, respectively. The risk of chromosome anomalies is around 2 percent. The risk of additional postnatal findings is low and the outcome in isolated cases is generally good.66, 67

CHAPTER 17

Prenatal Diagnosis of Fetal Malformations by Ultrasound 657

(a)

(b)

Figure 17.10 (a) Transverse and (b) sagittal plane of Blake’s pouch cyst in a fetus at 19.2 weeks of gestation.

Vermian hypoplasia

In vermian hypoplasia of Dandy–Walker variant, the vermis is formed but smaller than normal. The rates of associated central nervous system and other structural anomalies are 56 percent and 49 percent, respectively. The risk of chromosome anomalies is around 3 percent. When isolated, neurodevelopmental outcome is normal.66, 67 Dandy–Walker malformation

The sonographic findings in Dandy–Walker malformation are an enlarged fossa posterior with hypoplasia of the cerebellar vermis, dilatation of the fourth ventricle and a persistent Blake’s pouch cyst (Figure 17.11). Dandy–Walker malformation is a nonspecific marker of chromosomal abnormalities, microdeletions, genetic syndromes, congenital infection, or teratogens. In general, the associated mortality rate is high and intellectual development among survivors is poor. Associated central nervous system and other structural anomalies were present in 61 percent and 43 percent, with ventriculomegaly being the most common finding in 31 percent. The prevalence of chromosomal abnormalities is 16 percent. Abnormal neurodevelopmental occurs in 58 percent.66, 67

Fetal face The detection rate of fetal facial anomalies in utero is increased when ultrasound examination of the face includes an analysis in the three traditional sonographic examination planes – sagittal, coronal,

Figure 17.11 Transverse plane of a fetus with a Dandy–Walker malformation.

and axial. The coronal plane provides the best view of the upper and lower lips. Additionally, the nose, cheeks, eyelids, lenses, and forehead can be visualized in this plane. In the transverse plane the orbits, nasal bones, mandible, palate, alveolus (tooth buds), and lips should be evaluated and in the sagittal plane the fetal profile, including the chin, forehead, nasal bone, and nasal bridge. Recently, it has been demonstrated that by following specific steps in the mid-sagittal section of the fetus, the secondary palate can be well demonstrated.69 Moreover, the fetal uvula can also be demonstrated and presents as an “equals sign” in both the transverse and mid-sagittal plane. The absence of this sign can be used to diagnose an isolated cleft palate, since the presence of the uvula and consequently visualization of the “equal sign” proves an intact palate.70 With the improvement in ultrasound

658 Genetic Disorders and the Fetus

techniques an integrated approach of 2D and 3D ultrasound has been suggested to evaluate the fetal face to rule out a cleft lip and or palate. The 3D/4D ultrasound can be used for screening for facial clefts but is mandatory and an integral part in cleft analysis. The multiplanar reconstruction mode allows the simultaneous analysis of the three reference planes.71 The surface-rendering mode can be used to identify facial dysmorphologies and clefts. Cleft lip and/or palate

Anomalies of the lip and palate vary greatly, from isolated unilateral cleft lip (CL) or palate (P), to large defects involving the bilateral lip, alveolus, and palate. The prevalence of cleft lip with or without cleft palate (CL ± P) is around 1 per 1,000 compared to a much lower prevalence of isolated cleft palate of 1.3 to 25 per 10,000.72, 73 The prenatal detection rate of CL ± P depends on the population screened and varies greatly from 9 percent to 100 percent in low-risk populations and from 60 percent to 100 percent in high-risk populations, with an average around 40 percent.74 With the introduction of a routine 20 weeks scan, prenatal detection rates increased significantly, however, and now reach 70–88 percent in skilled hands.74–77 This impact could mainly be explained by the significant increase in prenatal detection rates of isolated CL ± P, since the prenatal detection rates of associated CL ± P were already high and remained stable.76, 77 Moreover, with the introduction of screening programs, the gestational age at the time of diagnosis also decreased gradually, allowing more time for additional testing.76, 77 The prenatal detection rate of isolated cleft palate remains low, however, between 0 and 22 percent,30 although some studies claim better results.74 In the case of micrognathia, the presence of a cleft palate should always be considered. When comparing the accuracy of the fetal anomaly scan in determining the oral cleft type, the postnatal findings were in accordance with the prenatal findings in 77 percent.78 Underestimation of the defect was more common than overestimation in 19 percent versus 4 percent, respectively.78 The amount of amniotic fluid may be helpful in recognizing the involvement of the palate in the case of CL ± P, since involvement of the palate was significantly more common in the presence

of polyhydramnios.79 Mistakes in differentiating unilateral as opposed to bilateral defects are rare.78 In the first trimester the prenatal diagnosis of CL ± P is more difficult but feasible. Thirty-five percent of the fetuses with a cleft lip and palate were diagnosed while the remaining 65 percent became apparent at the 20 weeks anomaly scan30 (Figure 17.12). Isolated cleft lip, however, was never diagnosed in the first trimester and 14 percent were only diagnosed after birth.30 Fetal facial anomalies can be isolated or associated with chromosomal anomalies or various multiple malformation conditions. In a systematic review a cleft lip had additional anomalies in only 13 percent versus 54 percent in fetuses with a cleft lip and palate.74 Although chromosome analysis is generally offered when CL ± P is found prenatally, the risk of chromosome anomalies is only 0.9 percent in isolated cleft lip versus 51 percent in associated cases.80 The most common chromosome anomaly found in isolated CL ± P is 22q11 microdeletion syndrome.81 In associated CL ± P, trisomy 18 is present in 56 percent of the fetuses, followed by trisomy 13 in 30 percent.80 In trisomy 13 the defect is frequently positioned in the midline of the face since it is often associated with holoprosencephaly. Besides chromosome anomalies, CL ± P may be present in many genetic syndromes. Micrognathia and retrognathia

Both micrognathia and retrognathia refer to an abnormal mandible and are an ominous sign.82 In micrognathia, the mandible is small, while in retrognathia it is displaced posteriorly. Most fetuses, however, will have a combination of both. Complete absence of the mandible, agnathia, is rare and may result in malposition or even fusion of the fetal ears at the level of the mandible.83 Both micrognathia and retrognathia may be diagnosed from the first trimester onwards in a mid-sagittal view, with the fetus showing an overbite (Figure 17.13). Care must be taken, however, not to misinterpret a long philtrum with micrognathia. The retronasal triangle view is also a useful technique to evaluate the fetal chin. This technique captures the coronal plane of the face in which the primary palate and the frontal processes of the maxilla are visualized simultaneously. Normal first-trimester fetuses display a characteristic gap between the right and left

CHAPTER 17

Prenatal Diagnosis of Fetal Malformations by Ultrasound 659

(a)

(b)

(c)

(d)

Figure 17.12 Fetus with a unilateral cleft lip and/or palate presenting at 19.2 weeks of gestation in transverse plane (a), coronal plane (b), and sagittal plane (c, d).

body of the mandible in this view (the “mandibular gap”).84 In the case of micrognathia, this gap is either absent or replaced by a bony structure. The majority of cases, however, will be detected in the second trimester.30 When isolated, micrognathia may be familial or part of the Pierre–Robin sequence, a craniofacial anomaly comprising mandibular hypoplasia, cleft secondary palate, and glossoptosis leading to life-threatening obstructive apnea and feeding difficulties during the neonatal period.85 When ear anomalies are present, Treacher Collins syndrome86 or hemifacial microsomia should be considered.87 Most of the fetuses with micrognathia have additional anomalies and will have underlying chromosome anomalies or genetic syndromes. Common chromosome anomalies are 22q11 microdeletion, trisomy 18 and 13, and triploidy.

Figure 17.13 Micrognathia demonstrated in a sagittal plane with the fetus showing an overbite.

660 Genetic Disorders and the Fetus

Pulmonary and thoracic abnormalities Congenital pulmonary airway malformation

Two of the most common long masses that may develop before birth are congenital pulmonary airway malformation (CPAM), previously known as congenital cystic adenomatoid malformation (CCAM) and bronchopulmonary sequestration (BPS). Neither is considered a genetic or hereditary condition, and both are unlikely to recur in a subsequent pregnancy. In a recent study the prenatal incidence of CPAM was 43 in 100,997 pregnancies.30 CPAM is a mass of abnormal fetal lung tissue that forms in pregnancy. Its exact cause is unknown, but it is hypothesized to be related to dysregulation of cell proliferation and cell apoptosis in the terminal bronchioles.88 CPAMs are classified sonographically as either macrocystic, with one or more cysts greater than 5 mm in diameter, or microcystic lesions, which appear as solid, echogenic masses89 (Figure 17.14). The mass may displace lung tissue and heart. BPS is a very similar condition, with a mass within the chest; however, it has a large vessel feeding it from the aorta. In general, CPAM is not visible on ultrasound during the first trimester, as this is a typical second-trimester diagnosis. However, an increased nuchal translucency (NT) was observed in 12 percent of all CPAM cases.30 Fetal hydrops may develop in large CPAMs because of compression of the heart and major

(a)

blood vessels in the thorax. The CCAM volumeto-head circumference ratio (CVR) is a tool that can be used to predict the development of hydrops, survival, need for fetal intervention, and the need for ventilation support or extracorporeal membrane oxygenation (ECMO), and length of hospital stay postnatally. It is calculated by dividing the volume of the mass (length × width × height × 0.52) by the head circumference.90 A CVR 1.6 is associated with adverse outcomes. When the CVR exceeds 1.6, steroid therapy can be considered.91 The effect of steroids should not be overestimated, as only small case series are present and no randomized trials exist. As in 20 percent of cases spontaneous regression of the CPAM occurs, especially in small, microcystic lesions, randomized trials are necessary to evaluate the value of steroid treatment.92 When hydrops develops, fetal intervention may be considered such as cyst aspiration or thoracoamniotic shunts. Postnatally, the management of CPAM or BPS is very similar. When symptomatic, there is little controversy that resection is indicated. When a lesion is asymptomatic there is greater debate regarding the benefit of resection versus continued observation.93 Arguments in favor of operation are decrease of pulmonary infections, poor diagnostic tools to distinguish a CPAM from a malignant pleuro-pulmonary blastoma, and better development of the remaining lung tissue after removal.94

(b)

Figure 17.14 Macrocystic congenital pulmonary airway malformation visible as a solid, echogenic structure in transverse plane (a) and sagittal plane (b).

CHAPTER 17

Prenatal Diagnosis of Fetal Malformations by Ultrasound 661

Congenital diaphragmatic hernia

Congenital diaphragmatic hernia (CDH) is a developmental discontinuity of the diaphragm that allows the abdominal organs to herniate into the chest. Although surgery can correct the diaphragmatic defect, in utero herniation of organs can result in pulmonary hypoplasia and pulmonary hypertension due to abnormal pulmonary vascular development and function. In a large study in 100,997 pregnancies 24 fetuses were diagnosed with CDH.30 An analysis of data from 31 population-based European registries revealed that there were 3,373 CDH cases reported among 12,155,491 registered births. Of 3,131 singleton cases, 353 (10 percent) were associated with a chromosomal anomaly, genetic syndrome or microdeletion and 784 (28 percent) were associated with other major structural anomalies. The total prevalence was 2.3 per 10,000 births for all CDH cases and 1.6 for isolated CDH cases.95 When cases resulting in termination of pregnancy and intrauterine fetal demise (IUFD) are added, the total incidence of CDH was 3.5 in 10,000 liveborn. The mortality rate between 1987 and 2013 was 36 percent: 44 percent during the first time period 1987–1999 and 27 percent in the later period 2000–2013.96 Prenatally, the diaphragm is imaged by ultrasonography as an echo-free space between the thorax and abdomen. CDH, which results from failure of closure of the posterolateral pleuro-peritoneal fold at 8–9 weeks of gestation, can be diagnosed by the demonstration of stomach and intestines (90 percent of the cases) or liver (50 percent) in the thorax and the associated mediastinal shift to the opposite side. Herniation is on the left in 80–85 percent of cases, on the right in 10–15 percent of cases, and bilateral in 45

15–25

Severe

20

5 mm110 (Figure 17.16). The prenatal incidence was found to be 8 in 100,997 pregnancies.30 This is a nonspecific finding in a wide variety of fetal and maternal

disorders, including hematologic, chromosomal, cardiovascular, renal, pulmonary, gastrointestinal, hepatic, and metabolic abnormalities, congenital infection, neoplasms, and malformations of the placenta or umbilical cord. Fetal hydrops can be of immune or nonimmune origin. In immune hydrops, the passage of maternal alloantibodies across the placenta cause fetal red cell destruction and in some cases bone marrow suppression, resulting in fetal anemia and subsequent hydrops. This disease process can affect both the fetus and neonate and is better labeled as hemolytic disease of the fetus and newborn (HDFN). Anemia and maternal alloantibodies are key in the diagnosis of immune hydrops fetalis. Hydrops is typically a second-trimester diagnosis and can be suspected from Doppler ultrasonographic assessment of the peak velocity of

(a)

(b)

(c)

(d)

Figure 17.16 Bilateral pleural effusion on a transverse plane of the fetal thorax (a), fetal abdomen (b), and fetal bladder with the umbilical arteries (c). (d) Coronal plane of hydrops in all compartments with clearly visible separation of the compartments by the diaphragm.

664 Genetic Disorders and the Fetus

systolic blood flow in the middle cerebral artery.111 Intravascular intrauterine erythrocyte transfusion can be offered to correct anemia. Livebirth rates as high as 95 percent have been reported in recent years,112 and when intrauterine transfusion is available, the outcome of fetuses with immune hydrops is just as good as nonhydropic fetuses with HDFN.113 With the widespread introduction of early alloantibody screening, immunoprophylaxis and the successful treatment of rhesus disease by fetal blood transfusions, lethal alloimmune hydrops has practically disappeared.113 Nonimmune fetal hydrops (NIHF) is the presence of ≥2 abnormal fetal fluid collections in the absence of red cell alloimmunization. NIHF is the cause in >85 percent of all affected individuals.114 The most common etiologies include cardiovascular, chromosomal, and hematologic abnormalities, followed by structural fetal anomalies, complications of monochorionic twinning, infection, and placental abnormalities (Table 17.2). Although in many instances the underlying cause may be determined by genetic screening, including WES, maternal antibody and infection screening, fetal

ultrasound scanning including echocardiography and Doppler studies, often the abnormality remains unexplained even after expert postmortem examination. In specific cases there are treatment options, like maternal transplacental administration of antiarrhythmic medication(s) for tachyarrhythmia, laser in twin-to-twin transfusion, intrauterine transfusion in anemia due to ParvoB19 infection, or a thoracoamniotic shunt in case of a chylothorax.115 The spontaneous resolution of hydrops has not been reported and the prognosis for this condition, irrespective of the underlying pathology, is extremely poor, with reported mortality rates of 50–95 percent.116, 117

Cardiovascular defects Congenital heart disease (CHD) is the most common congenital malformation. Gross structural abnormalities of the heart, or of major blood vessels that could actually or potentially affect the proper functioning of the heart are found in 8 per 1,000 livebirths118, 119 (see Chapter 1). The majority of the CHD is compatible with life, while

Table 17.2 Etiologies of nonimmune hydrops fetalis (before introduction of whole-exome sequencing). Etiology

Percent

Cause

Cardiovascular

17–35

Increased central venous pressure

Chromosomal

7–16

Cardiac anomalies, lymphatic dysplasia, abnormal myelopoiesis

Hematologic

4–12

Anemia, high output cardiac failure; hypoxia (α-thalassemia

Infectious

5–7

Anemia, anoxia, endothelial cell damage, and increased capillary

Thoracic

6

Vena caval obstruction or increased intrathoracic pressure with

Twin–twin transfusion

3–10

hypervolemia and increased central venous pressure

Urinary tract

2–3

Urinary ascites; nephrotic syndrome with hypoproteinemia

Gastrointestinal

0.5–4

Obstruction of venous return; gastrointestinal obstruction and

permeability) impaired venous return

infarction with protein loss and decreased colloid osmotic pressure Lymphatic dysplasia

5–6

Impaired venous return

Tumors including chorioangioma

2–3

Anemia, high output cardiac failure, hypoproteinemia

Skeletal dysplasia

3–4

Hepatomegaly, hypoproteinemia, impaired venous return

Syndromic

3–4

Various

Inborn errors of metabolism

1–2

Visceromegaly and obstruction of venous return, decreased

Miscellaneous

3–15

Unknown

15–25

erythropoiesis and anemia, and/orhypoproteinemia

Source: Society for Maternal-Fetal Medicine, Norton ME, Chauhan SP, Dashe JS. Society for maternal-fetal medicine (SMFM) clinical guideline #7: nonimmune hydrops fetalis. Am J Obstet Gynecol 2015;212(2):127. © 2015, Elsevier.115

CHAPTER 17

Prenatal Diagnosis of Fetal Malformations by Ultrasound 665

between 20–30 percent is severe, defined as being potentially life-threatening and requiring surgery within the first year of life.120–122 The cause of heart defects is heterogeneous and probably depends on the interplay of multiple genetic and environmental factors (see Chapter 1). Environmental factors that have been implicated in the causation of cardiac defects include maternal diabetes mellitus or collagen disease, exposure to drugs, and viral infections such as rubella. When a previous sibling has had a congenital heart defect, in the absence of a known genetic syndrome, the risk of recurrence is 2–3 percent. Specialist echocardiography in these cases at around 20 weeks of gestation can identify most critical cardiac defects. Since the majority of CHD, however, will occur in fetuses from women with no prior risk factor, the major challenge in prenatal diagnosis is to identify the high-risk group for referral to specialist centers and fetal echocardiography. When screening for CHD the cardiac screening examination should include both the four-chamber and outflow tract views.123 A normal heart is situated on the left side and occupies around one third of the area in the thorax. The cardiac axis points to the left by about 45 ± 20 degrees (2 SD). Normal heart rhythm rate lies between 110 and 160 beats per minute, but mild transient bradycardia may be observed when scanning the fetus. When assessing the four-chamber view one should evaluate if both atria and ventricles are equal in size, with the right side showing a moderator band. In the left atrium the foramen ovale flap should be seen and the entrance of the pulmonary veins. The cardiac crux should be intact as well as the ventricular septum. The offset of the right atrioventricular valve is slightly lower than the left. Views of the left and right ventricular outflow tracts should be an integral part of the fetal cardiac screening examination. To achieve this, angulation of the transducer from the four-chamber view is necessary for the demonstration of the normal aortic and pulmonary arterial origins from the left and right ventricles, respectively, to assess the normal size of the vessels and for the diagnosis of great arterial malalignment. When a fetal abnormality is suspected and women are referred for fetal echocardiography, pulsed Doppler studies for measurement of blood velocity across the valves should be added to the evaluation of the fetal heart,

since they are helpful in the assessment of fetal cardiac function (see Chapter 14). Moreover, in the setting of a fetal medicine unit, real-time directed M-mode echocardiography improves the accuracy of the cross-sectional scan in the diagnosis of cardiac defects and provides additional information on cardiac geometry and function. This method is particularly useful in the diagnosis and evaluation of dysrhythmias and the monitoring of in utero antiarrhythmic therapy. With the advancement in technology, assessment of the position of the fetal heart and the four-chamber view is now an integrated part of the first-trimester anomaly scan.123 In addition, since there is a greater incidence of cardiac anomalies among fetuses with a large NT,124, 125 tricuspid regurgitation,126 and abnormal ductus venosus,127 these findings in the first trimester warrant fetal echocardiography. These first-trimester markers subsequently lead to an increased number of fetuses with cardiac anomalies being diagnosed. Over the years prenatal detection rates have significantly increased. When national screening programs were first introduced and screening was primarily based on examination of the four-chamber view of the heart at the 20 weeks scan, only about 25 percent of the critical cardiac defects were prenatally diagnosed.128 The last decade detection rates in a low-risk population have slowly increased with the highest reported rate now being 60 percent,129 which is remarkably high in an unselected population, compared to similar programs that never exceeded 45 percent.95, 128, 130–132 For isolated CHD, however, detection rates are relatively low, not exceeding 44 percent.129 The detection rate very much depends on the severity of the anomaly, with CHD affecting the four-chamber view, such as hypoplastic left heart syndrome, being more often diagnosed prenatally compared to smaller defects, such as ventricular septal defects (Figure 17.17). The recognition that the introduction of the three-vessel view would significantly contribute to the prenatal detection of CHD had a dramatic impact.133 In a recent study it was demonstrated that the prenatal diagnosis of transposition of the great arteries (TGA) increased from 44 percent in the period without inclusion of the three-vessel view to 82 percent in the period including the

666 Genetic Disorders and the Fetus

Figure 17.17 A fetus with hypoplastic left heart syndrome.

three-vessel view. For tetralogy of Fallot the detection rate increased from 44 percent to 68 percent, respectively.134 With improved expertise and equipment, basic echocardiography for the examination of the four-chamber view, the connection of TGA, and the detection of dysrhythmias should be incorporated in the routine ultrasound screening programs for all pregnancies. Suspected anomalies can then be referred to specialized centers for further elucidation of the problem. Even in the first trimester, fetal echocardiography can pick up the majority of major CHD.135 When isolated, some of the cardiac defects resolve spontaneously (e.g. ventricular septal defect), some are correctable and carry intermediate operative risks (e.g. TGA), while others carry high operative risks (e.g. truncus arteriosus and left hypoplastic heart syndrome). Cardiac anomalies

Depending on the type of defect, CHD is more or less associated with other anomalies. TGA is generally isolated and carries a good prognosis, while tetralogy of Fallot and double outlet right ventricle are associated with chromosome anomalies and genetic syndromes. The presence or absence of other structural anomalies significantly adds to the risk of a more generalized problem. When other anomalies are present, CHD especially has a high risk of chromosome anomalies with a reported incidence around 20 percent, the most common being trisomy 21, trisomy 18, and 22q11 microdeletions.136, 137 Overall, it is advocated to offer invasive prenatal testing for fetal karyotyping or QF-PCR or FISH

and chromosomal microarray analysis whenever CHD is detected in a fetus, irrespective of how small or large the defect is. In a recent systematic review, array comparative genomic hybridization (array CGH) yielded additional clinically valuable information in 7.0 percent of fetal CHD cases, even after karyotyping and 22q11 FISH analysis were normal.138 More recently, WES has been added to the prenatal workup in some settings.139, 140 Cardiac anomalies can be the stimulus for a detailed search of anomalies; the opposite is also true, since in most genetic syndromes, other anomalies are likely to be detected first and only after a thorough evaluation of the fetal heart is a cardiac anomaly identified. Extracardiac defects, particularly craniospinal, gastrointestinal, and renal, are found in approximately one-third of cases. Based on the combination of anomalies, certain chromosome anomalies or genetic syndrome should be considered and rapid aneuploidy testing and chromosome microarray analysis should be part of standard care.141 However, since many genetic syndromes remain undetected with these tests, addition of WES had a positive effect on making a definite diagnosis.142 Ventricular septal defect

A ventricular septal defect (VSD) is a defect in the ventricular septum resulting in communication between the ventricular cavities (Figure 17.18). A VSD usually occurs as an isolated anomaly, but may also be present in TGA and tetralogy of Fallot. A recent study investigating the incidence and nature of abnormal chromosomal microarray analysis (CMA) results in a large cohort of pregnancies with VSD, showed that the rate of abnormal CMA findings (1.4 percent) in isolated VSD did not differ from that in pregnancies without a VSD. Atrioventricular septal defect

Atrioventricular septal defects (AVSDs) are characterized by a defect of the septal crux immediately above and below the atrioventricular valves (Figure 17.19). Coarctation of the aorta is the most frequent associated cardiac anomaly, while in 75 percent these defects are associated with other cardiac malformations.143 In cases of prenatally diagnosed AVSD up to 44 percent of the fetuses will be affected with Down syndrome.144

CHAPTER 17

Prenatal Diagnosis of Fetal Malformations by Ultrasound 667

(a)

(b)

Figure 17.18 Demonstrating a ventricular septal defect in transverse plane with (a) and without (b) color Doppler flow.

(a)

(b)

Figure 17.19 Demonstrating an atrioventricular septal defect with the atrioventricular valves during diastole (a) and systole (b). The septum primum is clearly missing.

Transposition of the great arteries

TGA is characterized by atrioventricular concordance and ventriculo-arterial discordance in which the aorta arises from the right ventricle and the pulmonary artery from the left ventricle. The diagnosis should be triggered when in the three-vessel view only two vessels are seen. Moreover, in three-vessel tracheal view a parallel course of great arteries may be observed. This cyanotic heart disease accounts for 5–7 percent of all CHDs and is usually an isolated finding.145 The prenatal diagnosis influences postnatal outcomes and therefore requires planned delivery and perinatal management (Figure 17.20). Tetralogy of Fallot

Tetralogy of Fallot consists of three main components that are the result of malalignment of the

infundibular septum: VSD, overriding aorta, and pulmonary stenosis (Figure 17.21). It occurs in 8–12 percent of fetuses with a cardiac anomaly.146 Sometimes the right ventricle may show hypertrophy. A right aortic arch is the most frequent cardiac anomaly in 22 percent. An abnormal cardiac angle will be noticed in 24 percent of fetuses with tetralogy of Fallot.147 Aneuploidy and 22q11 microdeletion are frequent associations.

Abdominal wall defects From approximately 7 weeks of gestation, the embryonic development of the intra-abdominal viscera can be followed by ultrasound.148, 149 At this gestation, the anterior abdominal wall is already

668 Genetic Disorders and the Fetus

(a)

(b)

Figure 17.20 Fetus with transposition of the great arteries presenting at 20.4 weeks of gestation. (a) Transverse plane of the normal four-chamber view. (b) Outflow tracts – transposition of the great arteries is clearly visible.

(a)

(b)

(c)

(d)

Figure 17.21 Outflow tract of the overriding aorta with ventricular septal defect (a, b), showing transverse plane at the level of the three-vessel view, presenting as the “U sign,” which corresponds to a right aortic arch, without and with color Doppler (c and d)).

CHAPTER 17

Prenatal Diagnosis of Fetal Malformations by Ultrasound 669

formed, but there is evidence of physiological herniation of the bowel into the cord: the abdominal cavity is too small to accommodate its contents. This physiological herniation is due to 90 degrees of counterclockwise rotation of the midgut and sonographically evident from 9 to 11 weeks of gestation. At approximately 12 weeks of gestation, the midgut will further rotate to 270 degrees and returns to the abdominal cavity. Beyond 12 weeks of gestation, midgut herniation is no longer physiologic.149 The liver is never present in physiological herniation, since it does not undergo physiologic migration outside the abdominal cavity. Omphalocele (exomphalos)

Omphalocele is an anterior abdominal wall defect with an incidence of 2 per 10,000 livebirths.150 The defect is herniated through the base of the umbilical cord, containing abdominal intestines like liver or spleen, and is covered by three layers: amnion, peritoneum, and Wharton’s jelly. Omphaloceles are divided into two groups: nonliver-containing or liver-containing omphalocele. Eighty percent of omphaloceles belong to the liver-containing group. Although not completely established, the pathogenesis of omphalocele is thought to have two mechanisms. The first one describes the failing of the 270 degrees counterclockwise rotation of the midgut back into the abdominal cavity. The second one is based on failure of closing of the lateral folds, resulting in a large abdominal wall defect through which abdominal content can herniate.151 Chromosomal and other associated structural anomalies are common in omphalocele. In the literature, associated structural anomalies are described in 35–70 percent of cases. Chromosomal anomalies are more prevalent in nonliver-containing omphalocele; approximately 60 percent are associated with fetal aneuploidy.152, 153 Trisomy 18, trisomy 13, Turner syndrome, and triploidy are the most reported chromosomal anomalies.154 Survival depends primarily on whether other malformations or chromosomal defects are present. In isolated cases, the malformation is correctable and survival rates of more than 90 percent are reported.155–157

First trimester Prenatal diagnosis of an omphalocele is made in 90 percent of cases and mostly in the first

trimester.30, 158 In a large prospective study with 44,859 pregnancies, fetal anomalies were detected in 488 cases and 213 were successfully identified through a first-trimester scan at 11+0 to 13+6 weeks of gestation, including all 104 cases of exomphalos, gastroschisis, megacystic, and body stalk anomaly.17 Since the liver is never a physiological finding outside the abdominal cavity, large, liver-containing omphalocele can be detected early in the first trimester, at approximately 9–10 weeks of gestation. Suggestive of liver is a homogeneous mass greater than 5–10 mm, present at the physiological herniation area.159, 160 The diagnosis of a nonliver-containing omphalocele is only possible after 12 weeks of gestation, when the physiologic midgut herniation has resolved.149 The diagnosis of omphalocele is made on the dorsal plane of the abdominal wall, demonstrating the herniated sac with its visceral contents and the umbilical cord insertion at the apex of the sac (Figure 17.22). Gastroschisis

Unlike omphalocele, gastroschisis is a paraumbilical abdominal wall defect in which the viscera herniate through the abdominal wall, usually located to the right side of the umbilical cord. Evisceration of the bowel occurs, sometimes in combination with other abdominal organs, floating freely in the amniotic fluid. Because the loops of intestines lie uncovered in the amniotic fluid, they become thickened, edematous, and matted. Several hypotheses have been proposed to explain the pathogenesis of gastroschisis, all

Figure 17.22 Large omphalocele at 13 weeks of gestation containing liver.

670 Genetic Disorders and the Fetus

involving defective formation or disruption of the body wall in the embryonic period with gut herniation. The latest hypothesis proposes that abnormal folding of the body wall results in a ventral body defect through which the gut herniates.161 The incidence of gastroschisis is increasing worldwide, approaching 1 in 4,000 live births in recent years.162 A consistent risk factor that has been reported in epidemiological studies is young maternal age. One European study found that compared with mothers aged 25–29, the relative risk was 7.0 (95 percent confidence interval 5.6–8.7) for mothers under 20 and 2.4 (95 percent CI 2.0–3.0) for mothers aged 20–24 years.163 Smoking, recreational drug use, and low BMI are also strongly associated with gastroschisis.162 It is usually an isolated finding and chromosomal anomalies are rare.158, 164 The prevalence of chromosomal anomalies in fetuses with isolated gastroschisis is not increased as compared to the normal population. Therefore, isolated gastroschisis is not a strong indication to pursue invasive diagnostics. However, other structural malformations are found in up to 16.6 percent of cases.165, 166 Over the past years, the postoperative survival rate increased from 75 percent to 96 percent and the incidence of complications decreased.166, 167

First trimester Most cases will be detected in first trimester. Like omphalocele, the prenatal detection rate is over 90 percent as a result of routine ultrasound screening.30 Diagnosis of gastroschisis is made

(a)

at the transverse plane, when demonstrating the normally situated umbilicus. It typically presents as a para-umbilical abdominal wall defect, to the right side of the midline, with herniation of free-floating bowel into the amniotic cavity168 (Figure 17.23).

Second and third trimester A fetus with gastroschisis will be monitored frequently because of fetal growth restriction that can occur in second or third trimester. Underestimation of the severity of the growth restriction is frequently seen, since measuring the abdominal circumference is complicated. Sonographers should also be alert to intra-abdominal bowel dilatation (Figure 17.24), gastric dilatation, and polyhydramnmios, since these findings are associated with bowel atresia.169 The number of cases of “sudden” intrauterine fetal death occurring in the last trimester of pregnancy is still significant (up to 13.6 percent). Usually the cause of death is not found170, 171 and occurs in both growth-delayed and nongrowth-delayed fetuses.170, 172 The hypothesis is that as a result of fetal hypo-albuminemia due to protein loss through the intestine, hypovolemia occurs with cardiovascular problems as a result.173 Regular fetal monitoring seems to decrease the intrauterine fetal deaths and results in premature birth. Body stalk anomaly

Body stalk anomaly is a severe and fatal condition with a reported incidence of 1 in 14,000 to

(b)

Figure 17.23 Gastroschisis in first trimester at 12.0 weeks of gestation. (a) Crown–rump length view of the fetus in mid-sagittal plane. (b) Gastroschisis in a transverse plane.

CHAPTER 17

Prenatal Diagnosis of Fetal Malformations by Ultrasound 671

(a)

(b)

Figure 17.24 (a) Transverse plane showing gastroschisis in the second trimester, detected at 19.3 weeks of gestation. (b) Coronal plane showing bowel dilation in the same fetus at 26.5 weeks of gestation.

(a)

(b)

Figure 17.25 Body stalk anomaly at 13 weeks of gestation. (a) Mid-sagittal plane with a large abdominal wall defect containing liver intestines and the bladder. (b) Severe kyphoscoliosis and a short umbilical cord.

1 per 31.000 pregnancies in large epidemiologic studies.155 It results from a developmental failure of the cephalic, caudal, and lateral body folds. In this severe abdominal defect the intrathoracic and abdominal organs lie outside the abdominal cavity, covered with a sac attached directly to the placenta.174 There is an absent or extremely short umbilical cord.175 Multiple additional defects may coexist, including skeletal, limb, and craniofacial abnormalities.176 Severe kyphoscoliosis is often present (Figure 17.25). Prenatal diagnosis is made after visualization of an anterior abdominal wall defect attaching the fetus to the placenta or uterine wall (Figure 17.26).

Bladder exstrophy and cloacal exstrophy

Bladder exstrophy is a defect of the caudal fold of the anterior abdominal wall. Whereas a small defect may cause epispadias alone, a large defect leads to exposure of the posterior bladder wall. Cloacal exstrophy is a severe multisystem congenital malformation involving the urinary, gastrointestinal, musculoskeletal, and neurological tracts. The disorder is also known as OEIS (omphalocele–exstrophy–imperforate anus–spinal dysraphism) complex.177 It most likely results from a very early defect in the closure of the ventral body wall rather than an abnormality related to premature rupture of the cloacal membrane.178

672 Genetic Disorders and the Fetus

(a)

(b)

(c) Figure 17.26 Body stalk anomaly at 10.3 weeks of gestation. (a, b) Mid-sagittal plane (a) and a transverse plane (b) showing the sac attached to the placenta. (c) Color Doppler showing the umbilical cord.

The incidence of bladder exstrophy is 1 per 20,000–40,000 with female predominance. Cloacal exstrophy is extremely rare, with a birth incidence of 1 per 200,000–400,000.178 A genetic basis is possible, but it is more likely to be a sporadic problem because it leads to sterility. Survival rates of 83–100 percent have been reported but involve technically challenging surgical management of the bowel and genitals. Although survival rates have improved due to advances in surgical reconstruction and techniques, the quality-of-life remains a concern.179 Accurate prenatal diagnosis of cloacal exstrophy appears to be less than 25 percent due to the rarity of the disorder.180 Major and minor criteria were defined in fetuses with cloacal exstrophy.180 Major criteria were nonvisualization of the urinary bladder, large midline infraumbilical anterior abdominal wall defect, omphalocele,

and meningomyelocele. Minor criteria were renal anomalies, lower extremity defects, and ascites.180 Prenatal ultrasound findings associated with bladder exstrophy are absence of bladder filling, low-set umbilicus, pubic bone diastasis, diminutive genitalia, and a lower abdominal mass that increases in size when the pregnancy progresses181, 182 (Figure 17.27). Prenatal detection is usually made in second trimester.182

Gastrointestinal anomalies On ultrasound, the fetal stomach is visible as a sonolucent structure in the upper left quadrant of the abdomen. A fluid-filled stomach should be detectable between 11 and 14 weeks of gestation, since the fetus begins to swallow amniotic fluid. The bowel is variably echogenic until the third trimester, when prominent meconium-filled loops

CHAPTER 17

Prenatal Diagnosis of Fetal Malformations by Ultrasound 673

Figure 17.27 Bladder exstrophy with nonvisualization of the urinary bladder.

Figure 17.28 The “pouch sign” in a fetus with esophageal atresia.

of large bowel are commonly seen. The normal esophagus is typically not visualized during ultrasound, it is collapsed and the amount of fluid is too small to be seen.

percent of cases, with trisomy 21 and trisomy 18 as most reported chromosomal anomalies.186 Prenatal diagnosis of esophageal atresia is poor and remains challenging, with reported detection rates between 23 percent to 32 percent.187 It should be suspected when fetal ultrasound fails to demonstrate the fetal stomach or a small stomach is visible, frequently in combination with a polyhydramnios. Ultrasound findings differ between types of esophageal atresia, depending on the presence of a fistula. In type A, without a fistula, the three key findings are: (i) polyhydramnios, (ii) absent stomach or a collapsed stomach, and (iii) blind-ending dilated upper esophageal pouch (“pouch sign”) (Figure 17.28). Prenatal diagnosis of esophageal atresia with a TEF is more difficult since the fistula allows the stomach to fill with amniotic fluid. In a systematic review of 1,760 affected fetuses, 77.9 percent of cases were correctly identified prenatally and only 21.9 percent of cases with a TEF.184 Ultrasound findings alone are thought to have a high false-positive rate for the diagnosis of esophageal atresia. Therefore, it is proposed that MRI should be used for establishing or ruling out a prenatal diagnosis of esophageal atresia.188

Esophageal atresia

Esophageal atresia refers to a congenitally interrupted esophagus and has an incidence of 2.4 in 10,000 births.183 Normally, during the 4th week of gestation, the primitive foregut separates into the trachea and the esophagus. Esophageal atresia arises from a developmental disruption of this separation. In 90 percent of esophageal atresia cases, a trachea-esophageal fistula (TEF) is present. Five types of esophageal atresia have been described in literature: type A – esophageal atresia without TEF (7 percent); type B – esophageal atresia with a TEF to the proximal esophageal segment (2 percent); type C – esophageal atresia with a TEF to the distal esophageal segment (86 percent); type D – esophageal atresia with TEF to both the proximal and distal esophageal segments (10 mm is always pathological and a reason for further evaluation. In the third trimester the cut-off for the diagnosis of ANH increases to 10 mm. Bilateral cases should have follow-up at higher frequency (Figure 17.31). In a recent study of 279 infants with secondtrimester isolated ANH (cut-off 5 mm), ANH had normalized (APPD 12 mm is seen in the first trimester the prognosis is extremely poor and parents often opt for termination of pregnancy.227 Posterior urethral valves are responsible for over 63 percent of LUTO, followed by urethral atresia or stenosis in 17 percent of cases.225 The male fetus is predominantly affected with posterior urethral valves, while in female fetuses urethral atresia is more common. Rare presentations of LUTO are megacystis–microcolon–intestinal hypoperistalsis syndrome (MMIHS),228 which may present with prune belly syndrome.229 When LUTO is suspected, ultrasound evaluation should include gender, associated anomalies such as heart (vertebral defects, anal atresia, cardiac

CHAPTER 17

Prenatal Diagnosis of Fetal Malformations by Ultrasound 679

defects, tracheo-esophageal fistula, renal anomalies limb anomalies [VACTERL association]); omphalocele (omphalocele exstrophy imperforate anus [OIES] complex); growth (overgrowth syndromes); and amniotic volume should be assessed. At the first-trimester scan, in over 30 percent of pregnancies with a megacystis, a trisomy was diagnosed,230 and in total 10 percent LUTO is associated with trisomy 21, 18, or 13.218 Moderate megacystis is in general more frequently associated with trisomies than severe megacystis.230 Although vesicoamniotic shunting seemed promising in treating LUTO caused by posterior urethral valves (PUV), thus far it seems to improve survival without improving renal function or decreasing the need for renal replacement therapy231 (see Chapter 29). Structural kidney malformations

Dysplastic kidneys Multicystic dysplastic kidneys

Multicystic dysplastic kidneys (MCDK) is a severe form of renal dysplasia in which the renal tissue is displaced by cysts. As a result the kidney consists of multiple noncommunicating cysts which differ in size and are separated by dysplastic parenchyma. Primitive nephrons filled with urine are the basis of the multiple cysts, which are seen initially at the periphery of the kidney. The kidney is enlarged and the overall shape is abnormal. The affected kidney is nonfunctional and frequently undergoes involution when the cysts shrink from absence of urine production. This leads to a picture similar to renal

(a)

agenesis. Unilateral MCDK is prevalent in 1:4,300 births, with the left kidney and males slightly more often affected232 (Figure 17.32). Bilateral MCDK occurs in 1:10,000 births. Typically the diagnosis of MCDK is made in the second trimester, but recent publications suggest that it may be visible by 2D and 3D ultrasound in the first trimester already233 (Table 17.4). Unilateral MCDK without additional anomalies is often sporadic,234 but bilateral MDCK should raise suspicion of aneuploidy or inherited conditions.218 Associated renal anomalies, often found in the contralateral kidney, include VUR and obstruction of the ureteropelvic junction, duplex kidney, agenesis, or pelvic kidney. The most common extrarenal abnormalities are heart defects, esophageal or intestinal atresia, spinal abnormalities, and the VACTERL association. Bilateral MDCK, formerly known as Potter type 2, can lead to “Potter sequence,” that is pulmonary hypoplasia secondary to impaired renal function and anhydramnios. A recent study in 53 cases of prenatally suspected MCDK, of which 46 cases were liveborn and confirmed post natally (38 survivors, 8 nonsurvivors) revealed that extrarenal anomalies, bilateral MCDK, contralateral renal anomalies, and anhydramnios were significantly associated with death or need for dialysis (all p < 0.0001).235

Polycystic kidney disease Polycystic kidney disease is typically due to terminal epithelial differentiation disruption. This includes autosomal recessive polycystic kidney

(b)

Figure 17.32 Unilateral multicystic dysplastic kidney demonstrated in transverse plane (a) and coronal plane (b).

680 Genetic Disorders and the Fetus

Table 17.4 Conditions associated with large and echogenic kidneys from Yulia and Winyard218 . Amniotic

Renal

APPKD

Renal

fluid

cysts

dilatation

size

Macrosomia

Extrarenal abnormalities

Inheritance

Obstruction

N/oligo

+/−

+

+



+/−

Sporadic

Dysplasia

N/oligo

+/−

+/−

+



+/−

10 AD

ARPKD

N/oligo





+





AR

ADPKD

N/oligo

+



+





AD

Beckwith–Wiedeman

N/poly



+/−

+

+

+

AD or disomy

Perlman

N/oligo



+/−

+

+

+

AR

+

+

+

X-linked

Meckel–Gruber

Oligo





+



+

AR

Nephrocalcinosis

N











Sporadic

Simson–Golabi–Behmel

ARPKD, autosomal recessive polycystic kidney disease; ADPKD, autosomal dominant polycystic kidney disease; AD, autosomal dominant; AR, autosomal recessive. Source: Yulia A, Winyard P. Management of antenatally detected kidney malformations. Early Hum Dev 2018;126:38. © 2018, Elsevier.218

disease (ARPKD), which occurs in approximately 1:20,000 births.236 It is characterized by multiple microscopic cysts in the distal collecting ducts. It is caused by mutations in the PKHD1 gene. Characteristic ultrasound findings are two large, hyperechogenic kidneys (Figure 17.33). The differential diagnosis includes Beckwith–Wiedeman syndrome, trisomy 13, or Meckel–Gruber syndrome. The timing and presentation may vary, but when anhydramnios is present, it is almost always a lethal condition. Formerly this was also referred to as infantile polycystic kidney disease or Potter 1 renal dysplasia. Autosomal dominant polycystic kidney disease (ADPKD), or adult polycystic kidney disease, is far more common with an incidence of 1:500–1,000 livebirths.237 Ultrasound findings are similar to ARPKD, with bilateral enlarged (hyperechogenic) kidneys with or without cysts. It can either be isolated or with cardiovascular and biliary tract involvement.238 In adulthood, ADPKD accounts for 5–10 percent of all end-stage renal disease. Symptoms with cysts formation and renal failure usually start in middle age, but prenatal presentation may occur in 2–5 percent of cases, mainly in the third trimester, and is associated with adverse outcomes. ADPKD is associated with mutations in the PKD1 or PKD2 genes.239

Nephronophthisis (former Potter 3 renal dysplasia) Nephronophthisis (NPH) is a renal ciliopathy and is characterized by reduced renal concentrating ability, chronic tubulo-interstitial nephritis, cystic renal disease, and progression to end-stage renal disease (ESRD).240 It includes autosomal recessive renal diseases in which multiple cysts are formed within the cortico-medullary region. Nephronophthisis is responsible for 2.4–15 percent of ESRD in children.241 Mutations can be found in one of the 19 known NPH-related genes involved in ciliary structure or function. Well-known nephronophthisis syndromes are Meckel–Gruber syndrome, characterized prenatally by hyperechogenic kidneys, polydactyly, and encephalocele, and Bardet–Biedl syndrome, characterized by hyperechogenic kidneys and polydactyly.242 Renal agenesis

Unilateral renal agenesis Unilateral renal agenesis, defined as congenital absence of one kidney, accounts for 5 percent of renal malformations243 (Figure 17.34). Unilateral renal agenesis occurs in 1 in 1,000 births. Although it may be secondary to a chromosomal abnormality or part of a genetic syndrome, such as Fraser syndrome, more commonly it is an isolated finding. It is diagnosed either antenatally or in a workup for a urinary tract infection. In nonsyndromic cases, the risk of recurrence is approximately 3 percent. However, 4.5 percent of first-degree relatives of affected

CHAPTER 17

Prenatal Diagnosis of Fetal Malformations by Ultrasound 681

(a)

(b)

Figure 17.33 Polycystic kidney disease presenting two large hyperechogenic kidneys in transverse plane (a) and sagittal plane (b).

Figure 17.34 Unilateral renal agenesis demonstrated in coronal plane.

Renal agenesis is accompanied by absence of both ureters and renal arteries, and results from failure of ureteric buds to either form or reach the metanephric mesenchyme which results in apoptosis.219 Anhydramnios around 18–20 weeks of gestation is the most striking feature on ultrasound that will lead to the diagnosis of bilateral renal agenesis. Color Doppler flow of the renal arteries will facilitate the detection of bilateral renal agenesis, especially when imaging due to oligo- or anhydramnios is difficult.216 If either unilateral of bilateral renal agenesis is identified, a detailed ultrasound of all other organ systems must be done because the risk of a genetic syndrome may be as high as 30 percent.242, 246

infants have unilateral renal agenesis themselves, against 0.3 percent in a control population.244 Unilateral renal agenesis is easily overlooked as amniotic fluid is normal due to hyperfiltration and compensatory hyperplasia of the contralateral kidney and normal bladder filling. Differential diagnosis includes, among others, a pelvic kidney.

Abnormalities of pelvic migration

Bilateral renal agenesis Bilateral renal agenesis complicates approximately 1 in 10,000 to 3 in 10,000 births.242 In general, it is a lethal condition, leading to pulmonary hypoplasia because of lack of amniotic fluid in the critical stage of lung development between 16 and 20 weeks of gestation.245 First-trimester diagnosis is possible, but more difficult as amniotic fluid may appear normal. However, the bladder is not filled with urine.

Pelvic kidney A pelvic kidney may be considered when the kidney cannot be visualized at its usual anatomical site, but is present in the pelvis. The diagnosis can easily be missed because the adrenal glands can be mistaken for kidneys. Its incidence is approximately 1 in 1,700 pregnancies.247 In most cases a pelvic kidney is isolated, but it has also been associated with genetic disorders, including Turner

Disruption of the normal embryologic migration of the kidneys can result in renal ectopia (pelvic kidney) and/or fusion abnormalities (horseshoe kidney). Most patients are asymptomatic but complications such as urinary tract infection or reflux occur.

682 Genetic Disorders and the Fetus

syndrome (45,X), Williams–Beuren syndrome, and Antley–Bixler syndrome.248–250 In most of these conditions, a pelvic kidney is not isolated and additional anatomic abnormalities are present. A recent study revealed no difference in copy number variants (CNVs) detected with chromosomal microarray analysis in fetuses with a pelvic kidney as compared to a normal population.251 Therefore invasive procedures solely for isolated pelvic kidneys can be questioned.

Horseshoe kidney A horseshoe kidney is seen in 1:400 persons. A horseshoe kidney is a result of fusion of the upper or lower poles of the kidneys before 4–6 weeks of embryological development and before the kidneys have migrated upward and rotated on their long axis.252

Skeletal anomalies There are a wide range of skeletal anomalies with different underlying pathologies such as skeletal dysplasia, idiopathic limb reduction, chromosome anomalies, teratogens, genetic syndromes, and metabolic conditions. Skeletal dysplasias are relatively rare, each with a specific mode of inheritance, genotype, phenotype, recurrence risk, and implications for neonatal survival and quality of life (see Chapter 20). In the past, skeletal anomalies were usually only recognized postnatally, but with the introduction of routine scanning, many may be discovered in the first trimester because of specific sonographic findings.253, 254 In the case of skeletal dysplasia it is important to at least differentiate between the lethal and nonlethal types. Knowledge on the specific prenatal features that are present in skeletal dysplasia, however, will greatly facilitate diagnosis. Gene discovery and WES (see Chapters 14 and 20) adds to an accurate prenatal diagnosis in additional cases. A definitive diagnosis facilitates parental counseling necessary to provide parents with accurate information regarding the prognosis and the option to make autonomous reproductive choices. When a skeletal anomaly is found during a routine ultrasound or a skeletal dysplasia is prenatally suspected, a systematic examination of all the skeletal structures and all the organ systems is recommended to arrive at the correct diagnosis.255

All limbs must be evaluated for length, shape, mineralization, and movement, and associated abnormalities in other systems, particularly the head, thorax, and spine, should be sought (see Chapter 20). A putative diagnosis may then allow definitive confirmation via mutation analysis. The majority of bones of the appendicular system can be imaged from the first trimester onwards, and several nomograms relating the length of long bones to gestational age or biparietal diameter have been published.256 For other skeletal structures, nomograms relating to the head, clavicles, mandible, and thorax are widely available.257–259 Prenatal assessment of the fetus suspected of a skeletal anomaly will usually start with measurement of all the long bones. The absence or degree of hypoplasia should be evaluated. Anomalies can be both symmetrical and asymmetrical and the pattern of shortening may be variable. Most often the trigger leading to fetal evaluation for skeletal dysplasia will be the finding of a short femur. It should be recognized that the most common cause of a short femur is a genetically small but healthy child, either due to being small for gestational age or due to fetal growth restriction (Table 17.5). The most frequent anomaly in fetuses with a short femur is Down syndrome. In the case of a short femur and the prenatal suspicion of achondroplasia, usually in the third trimester because of a sudden decline in femoral growth velocity, the proximal diaphysis–metaphysis angle of the femur should be measured. From 20 weeks onwards, this angle is generally wider in fetuses with achondroplasia as compared to fetuses affected by intrauterine growth restriction (IUGR), small for gestational age (SGA) fetuses, and normal fetuses, and exceeds 130 degrees260, 261 (Figure 17.35). Severe lethal skeletal dysplasia such as osteogenesis imperfecta type II, achondrogenesis, thanatophoric dysplasia, and diastrophic dysplasia, have a first-trimester detection rate of 70 percent.30 In the case of achondroplasia, however, the diagnosis may not become obvious until the third trimester because of a sudden decline in femoral growth velocity.262 When examining the bones it is also important to assess whether bowing, fractures, or ephiphyseal stippling is present. A minor degree of lateral curvature of the femur is commonly seen in normal fetuses. Fetuses with achondroplasia may have mildly bowed femora.262

CHAPTER 17

Prenatal Diagnosis of Fetal Malformations by Ultrasound 683

Table 17.5 Differential diagnosis in short femur. Feature

SGA

FGR

Down syndrome

Achondroplasia

Ultrasound anomalies

None

None

Markers AVSD

Frontal bossing

Femur length