Approach to Intellectual Disability

Similar documents
Approach to the Child with Developmental Delay

Approach to Mental Retardation and Developmental Delay. SR Ghaffari MSc MD PhD

Neonatal Hypotonia Guideline Prepared by Dan Birnbaum MD August 27, 2012

Neonatal Hypotonia. Encephalopathy acute No encephalopathy. Neurology Chapter of IAP

Sharan Goobie, MD, MSc, FRCPC

Medical Conditions Resulting in High Probability of Developmental Delay and DSCC Screening Information

CURRENT GENETIC TESTING TOOLS IN NEONATAL MEDICINE. Dr. Bahar Naghavi

22q11.2 DELETION SYNDROME. Anna Mª Cueto González Clinical Geneticist Programa de Medicina Molecular y Genética Hospital Vall d Hebrón (Barcelona)

Intellectual Disability

AMERICAN BOARD OF MEDICAL GENETICS AND GENOMICS

Approach to the Genetic Diagnosis of Neurological Disorders

The child with hemiplegic cerebral palsy thinking beyond the motor impairment. Dr Paul Eunson Edinburgh

New and Developing Technologies for Genetic Diagnostics National Genetics Reference Laboratory (Wessex) Salisbury, UK - July 2010 BACs on Beads

Original Policy Date

Corporate Medical Policy

ESP 755A SUMMER Multiple Choice Identify the choice that best completes the statement or answers the question. 1. Autosomal recessive disorders

GENETICS ROTATION OBJECTIVES MATERNAL-FETAL MEDICINE FELLOWSHIP

The University of Arizona Pediatric Residency Program. Primary Goals for Rotation. Genetics

Summary. Syndromic versus Etiologic. Definitions. Why does it matter? ASD=autism

Neonatal Seizure. Dr.Nawar Yahya. Presented by: Sarah Khalil Zeina Shamil Zainab Waleed Zainab Qahtan. Supervised by:

Supplementary Online Content

Genetic Testing 101: Interpreting the Chromosomes

Unifactorial or Single Gene Disorders. Hanan Hamamy Department of Genetic Medicine and Development Geneva University Hospital

Babies First and CaCoon Risk Factors (A Codes and B Codes)

Cerebral Palsy. What is Cerebral Palsy? Clues to Diagnosis of Cerebral Palsy 12/30/2012

Evolution of Genetic Testing. Joan Pellegrino MD Associate Professor of Pediatrics SUNY Upstate Medical University

Childhood Hearing Clinic causes of congenital hearing loss Audit of results of investigations

Atlas of Genetics and Cytogenetics in Oncology and Haematology

What s New in Newborn Screening?

Developmental delay Poor School Progress. I Smuts Department of Paediatrics

Movement disorders in childhood: assessment and diagnosis. Lucinda Carr

What s the Human Genome Project Got to Do with Developmental Disabilities?

Clinical evaluation of microarray data

RACP Congress 2017 Genetics of Intellectual Disability and Autism: Past Present and Future 9 th May 2017

Challenges of CGH array testing in children with developmental delay. Dr Sally Davies 17 th September 2014

MICROCEPHALY DEVELOPMENTAL IMPLICATIONS IN BIRTH-3 Diana M. Cejas, MD, MPH Child Neurology Fellow The University of Chicago Comer Children s Hospital

Presentation and investigation of mitochondrial disease in children

Prenatal Prediction of The Neurologically Impaired Neonate By Ultrasound

Dysmorphology And The Paediatric Eye. Jill Clayton-Smith Manchester Centre For Genomic Medicine

Egypt 90 Million People Power Seven Thousands Year Culture 29 Governorates

6 AAMD Classifications

Genetics and Developmental Disabilities. Stuart K. Shapira, MD, PhD. Pediatric Genetics Team

Genetics of Congenital Heart Diseases by WINCARS Association

Fetal Medicine. Case Presentations. Dr Ermos Nicolaou Fetal Medicine Unit Chris Hani Baragwanath Hospital. October 2003

Chapter Objectives 9/21/2017. Chapter 6 Biological Aspects and the Promises of Prevention. 8 points possible

NYEIS Version 4.3 (ICD) ICD - 10 Codes Available in NYEIS at time of version launch (9/23/2015)

Epilepsy in children with cerebral palsy

Lab #10: Karyotyping Lab

Genetics and Genomics: Applications to Developmental Disability

Case 1B. 46,XY,-14,+t(14;21)

Intellectual Disability. Dr Catherine Marraffa RCH April 2014

Neurodevelopment. Checkpoints in children with congenital heart disease

NEONATAL SEIZURE. IAP UG Teaching slides

Applications of Chromosomal Microarray Analysis (CMA) in pre- and postnatal Diagnostic: advantages, limitations and concerns

GENOTYPE-PHENOTYPE CORRELATIONS IN GALACTOSEMIA COMPLICATIONS COMPLICATIONS COMPLICATIONS LONG-TERM CHRONIC COMPLICATIONS WITH NO CLEAR CAUSE

Multiple Copy Number Variations in a Patient with Developmental Delay ASCLS- March 31, 2016

Common Genetic syndromes. Dr. E.M. Honey Department Genetics Division Human Genetics University of Pretoria

CHROMOSOMAL MICROARRAY (CGH+SNP)

MEDICAL GENETICS CLINICAL CARE ROTATION

SEX-LINKED INHERITANCE. Dr Rasime Kalkan

Neurology. Access Center 24/7 access for referring physicians (866) 353-KIDS (5437)

Learning Outcomes: The following list provides the learning objectives that will be covered in the lectures, and tutorials of each week:

Faravareh Khordadpoor (PhD in molecular genetics) 1- Tehran Medical Genetics Laboratory 2- Science and research branch, Islamic Azad University

Early Identification of Young Children with Deaf-Blindness

Childhood epilepsy: the biochemical epilepsies. Dr Colin D Ferrie Consultant Paediatric Neurologist Leeds General Infirmary

Exploding Genetic Knowledge in Developmental Disabilities. Disclosures. The Genetic Principle

What s New in Newborn Screening?

Index. Child Adolesc Psychiatric Clin N Am 16 (2007) Note: Page numbers of article titles are in boldface type.

Genetic diagnosis in clinical psychiatry: A case report of a woman with a 47,XXX karyotype and Fragile X syndrome

Prenatal Diagnosis: Are There Microarrays in Your Future?

EGI Clinical Data Collection Form Cover Page

Evaluation of the Hypotonic Infant and Child

SELECTIVE VULNERABILITY (HYPOXIA AND HYPOGLYCEMIA)

Birth mother Foster carer Other

A. Definitions... CD-157. B. General Information... CD-158

The high risk neonate

Screening, assessment and management of neonates and infants with complications associated with Zika virus exposure in utero

intracranial anomalies

Genetic Conditions and Services: An Introduction

CLINICAL GENETICS 1. GENERAL ASPECTS OF THE SPECIALISATION. 1.1 Purpose of the specialisation

Syndromic X Linked Mental Retardation

A Lawyer s Perspective on Genetic Screening Performed by Cryobanks

This fact sheet describes the condition Fragile X and includes a discussion of the symptoms, causes and available testing.

Cerebral Malformation gene panel

Difficulties at Birth: Long Term Developmental Outcomes

The University of Arizona Pediatric Residency Program. Primary Goals for Rotation. Neurology

Central Nervous System

Pearls in Child Neurology. Edgard Andrade, MD, FAAP Assistant Professor University of Florida

Red flags for clinical practice - guidance on indicators that your patient may have a genetic condition

Section F: Discussing the diagnosis and developing a management plan

chromosomal anomalies and mental pdf Chapter 8: Chromosomes and Chromosomal Anomalies (PDF) Chromosomal abnormalities -A review - ResearchGate

Child Neurology Elective PL1 Rotation

NEONATAL MUSCULAR HYPOTONIA

CLINICAL AUDIT SUMMARY CLINICAL AUDIT SUMMARY. Diagnosis and Recognition of Congenital Cytomegalovirus in Northern Ireland

UNIT IX: GENETIC DISORDERS

Unusual Modes of Inheritance. Wayne Lam

NEONATAL SEIZURES-PGPYREXIA REVIEW

Neonatal manifestations of lysosomal storage diseases

The Floppy Baby. Clare Betteridge

New Patient Information Form

Transcription:

Approach to Intellectual Disability Dr Prajnya Ranganath Definition Significant sub-average intellectual function existing concurrently with deficits in adaptive behaviour and manifested during the developmental period. Intellectual disability/ mental retardation has 3 components: i. Significantly abnormal intellectual performance (determined by a test of intelligence) ii. iii. Impairment of the ability to adapt to the environment Onset during development before the age of 18 years Developmental delay is a term usually used for young children especially 5 years in whom formal IQ testing is not possible, refers to delay in attainment of milestones and implies deficit in learning and adaptation. When all spheres are involved (gross & fine motor, social & adaptive, language), referred to as global developmental delay. Mental retardation/ intellectual disability includes: Static non - progressive disorders with onset of cognitive impairment from birth/ early childhood Progressive disorders with onset of cognitive impairment in childhood Hereditary neurodegenerative and metabolic disorders with neuro-regression beginning some time after a period of normal development Mental retardation/ intellectual disability is said to be present when the intelligence quotient (IQ) is less than 70. Classification based on IQ: Mild: 50-70 Moderate: 35 50 Severe: 20 35 Profound: < 20

Etiology Known Genetic Causes: o Chromosomal 30 35% Down syndrome Other chromosomal anomalies Microdeletion syndromes o Single gene disorders 8 10% Multiple malformation syndromes 4 5% Fragile X and other X linked MR 1 3% Inborn errors of metabolism 4% Multiple malformation syndromes: 15% (non monogenic) o Known sporadic syndromes o New or private syndromes Central nervous system malformations: 5 7% o Neural tube defects o Hydrocephalus o Neuronal migration disorders o Microencephaly o Other structural anomalies: holoprosencephaly, Dandy Walker malformation etc. CNS dysfunction due to identified 15% perinatal, prenatal & postnatal causes o Infection fetal infection, postnatal meningitis or encephalitis o Teratogenic exposures o Asphyxia o Hemorrhage or infarction Static insults presenting as cerebral palsy: 5 10% without substantial evidence of an environmental cause Unidentified: 40 60%

Down syndrome is the most common genetic cause of mental retardation. It affects 1 in 700 800 live born children and accounts for 20-30% of all cases of MR. Trisomy 21 is found in 95% cases of Down syndrome, translocation involving chromosome 21 occurs in 3 4% cases and mosaic trisomy 21 occurs in 1% of cases. Fragile X syndrome is the most common monogenic disorder associated with MR and is the most common inherited cause for MR. It accounts for 1 2 % of all cases of MR and has an X linked inheritance pattern. Diagnostic Approach to Intellectual Disability 1. Detailed history 2. Physical examination 3. Baseline hematological and biochemical investigations 4. Thyroid function tests 5. Neuroimaging 6. Karyotyping 7. Fluorescence in situ hybridization (FISH)/ Multiplex probe ligation dependent amplification (MLPA) study for specific suspected microdeletion syndromes 8. Fragile X molecular genetic testing 9. Molecular genetic testing/ metabolic testing for suspected specific monogenic conditions 10. Cytogenetic microarray study History Antenatal period (age at conception, teratogens [drugs and infection], other maternal illnesses, abnormal ultrasound) Perinatal period (prematurity, diificult labour, asphyxia, growth retardation, sepsis, hyperbilirubinemia) Postnatal period (seizures, history of meningitis /encephalitis, abnormal posturing, spasticity, visual and hearing problems, exposure to lead, behavioral changes, deterioration in school performance) Detailed developmental history (time of attainment of milestones; regression of milestones)

History suggestive of metabolic disorders (failure to thrive, recurrent unexplained illness, seizures, ataxia, loss of psychomotor skills, recurrent somnolence/ coma, lethargy, abnormal body odour) Family history Detailed family history including 3 generation pedigree Family history of MR, developmental delay, seizures and unexplained psychiatric disorders. Try to ascertain the pattern of inheritance History of congenital malformations, miscarriages, stillbirths, and early childhood deaths. Physical examination Complete anthropometric assessment (height, weight, head circumference, upper segment: lower segment ratio) Detailed dysmorphologic evaluation from head to toe and comparison of findings with available literature, OMIM and dysmorphology databases (such as London Dysmorphology DataBase, POSSUM) Detailed neurological evaluation: o higher mental functions, o cranial nerve deficits, o motor and sensory system examination, o cerebellar signs, o involuntary/ abnormal movements, o persistence of neonatal reflexes Ophthalmological examination should be done in all cases to look for corneal, lenticular and retinal abnormalities can provide an important clue to the diagnosis o Retinal changes/ pigmentary degeneration: specific syndromes (Norrie disease, Bardet Biedl syndrome, mitochondrial disorders, NCL) o Chorioretinitis: congenital intrauterine infections o Cherry red spot: specific storage disorders (gangliosidosis, mucolipidosis, Niemann Pick disease) o Corneal clouding: mucopolysaccharidosis, oligosaccharidosis, mucolipidosis

o Cataract: galactosemia, congenital rubella syndrome, Lowe syndrome, congenital myotonic dystrophy, Cockayne syndrome o Lens dislocation: homocystinuria, sulfite oxidase deficiency Assessment of the behavioural phenotype (e.g. self mutilation in Lesch Nyhan syndrome, self mutilation, aggression and sleep disturbances in Smith Magenis syndrome, autistic features in Fragile X and duplication 15q, hyperphagia and sleep disturbances in Prader Willi syndrome, typical hand movement stereotypes and breathing abnormalities in Rett patients) Complete general and systemic examination Baseline Investigations: Complete hemogram including RBC indices (may provide clue to diagnosis eg. alpha thalassemia mental retardation syndrome, macrocytosis in abnormalities of vitamin B12 metabolism etc.) SGPT: gross liver function assessment Serum creatinine: gross renal function assessment Serum creatine phosphokinase: young male patients with DMD may initially present with developmental delay Serum T4 & TSH: Hypothyroidism must be excluded in all cases of developmental delay especially when clinical features of hypothyroidism are seen and in younger children (< 2 years) where no specific cause is identifiable Hypothyroidism may be associated with some genetic syndromes e.g. Down syndrome Very important to exclude because it is a treatable cause. Neuroimaging: Recommended when : o abnormal head size: microcephaly/ macrocephaly o seizures o focal motor/ neurological findings on neurological examination o associated malformations esp. facial malformations

Preferable to do if no other aetiology is obvious; in conditions like neuronal migration anomalies and some cases of pre/ perinatal insult there may be no other clinical diagnostic clue MRI is preferable to CT Scan for neuro-imaging and is much more informative Neuroimaging helps to identify hypoxic ischemic sequelae, vascular insults, neuroectodermal syndromes, intracranial structural defects and neuronal migration abnormalities. CT Scan head is better for documentation of intracranial calcifications associated with old hemorrhage, tuberous sclerosis complex and congenital CMV or toxoplasmosis infection, and for craniosynostosis Specific tests based on the clinical features in each individual case: Examples: Electrophysiological studies: EEG/ Audiometry/ EMG/ NCV TORCH serology: for suspected congenital intrauterine infections in infants Ultrasonography to look for renal malformations Echocardiography to look for cardiac malformations Genetic Evaluation: Should be done in all cases of intellectual disability without a definitely proven environmental cause. The label of cerebral palsy due to adverse perinatal events should not be given until there is definite evidence in neuroimaging. Karyotyping: Should be done in all cases with intellectual disability, where a specific monogenic cause has not been clinically identified Conventional karyotyping (up to 550 band-level) yields a diagnosis in 2.5 3% of unexplained/ idiopathic MR cases FISH/ MLPA study: To confirm the diagnosis of clinically identified microdeletion syndromes such as Prader-Willi syndrome, Angelman syndrome, Di George syndrome, Williams syndrome etc. MLPA panels to test for multiple common microdeletions in one test are available commercially Testing for Fragile X syndrome:

Fragile X screening can be done through PCR- based tests but confirmation requires Southern Blot testing. If fragile X molecular genetic testing is done using a phenotypic checklist of 7 features (proposed by de Vries et al, J Med Genet 1999) the diagnostic yield is ~7 8 % (long jaw, high forehead, large and/or protuberant ears, hyper extensible joints, soft and velvety palmar skin with redundancy on the dorsum of the hands, testicular enlargement, and behavior of initial shyness and lack of eye contact followed by friendliness and verbosity). Younger children have subtle dysmorphism which may be missed; therefore fragile X testing is recommended in all children especially males with idiopathic MR even in the absence of a family history. Molecular genetic testing: If a specific monogenic disorder is suspected clinically, mutation analysis of the relevant gene to be done for confirmation Metabolic testing: If a specific inborn error of metabolism (IEM) is suspected clinically, the relevant biochemical assay such as enzyme assay/ plasma amino acid HPLC/ urine GCMS for organic acids etc. is to be done for confirmation. Most IEMs present with neuroregression and have a progressive course Routine metabolic evaluation in unexplained MR has an average diagnostic yield of only around 1%. Cytogenetic microarray: Has a diagnostic yield of around 12 15% in idiopathic MR. Has been recommended as the first line diagnostic test for idiopathic MR by the American College of Medical Genetics. Detects genome-wide copy number variations (CNVs- deletions/ duplications). Algorithm has to be followed to determine the pathogenicity of a detected CNV. Management: For most cases of MR, treatment is mainly symptomatic and supportive. Mainstay of therapy: early stimulation/ physiotherapy/ vocational training/ special schooling/ speech therapy Institutionalization is to be discouraged and integration into society is encouraged. Multidisciplinary approach: for appropriate management

Genetic counseling Depends on the aetiology: A. Chromosomal causes of MR: usually not familial except when one of the parents is a carrier of a balanced chromosomal rearrangement. risk of recurrence in de novo chromosomal disorders is low (usually < 1%) In translocation Down syndrome, when one of the parents is a balanced translocation carrier the recurrence risk is variable: - 2-5% if the father is a carrier - 10 15 % if the mother is a carrier - 100% if either parent is a carrier of 21; 21 translocation B. Single gene disorders: Risk of recurrence depends on the mode of inheritance: Autosomal recessive disorder: the risk of recurrence is 25% in the next sibling Autosomal dominant disorder: the risk of recurrence for the sibling is 50% if one of the parents is affected; in a sporadic case (de novo mutation), the risk of recurrence in the sibling is very low but gonadal mosaicism cannot be ruled out. X-linked recessive disorders: the risk of recurrence for boys is 50%, whereas females usually do not manifest the disorder. C. Environmental causes: Recurrence is unlikely but can occur if the causative agent persists in the environment of the other child e.g. lead exposure, intrauterine exposure to teratogens like alcohol or specific drugs D. When no etiology is identified: Empiric risks of recurrence can be predicted when no specific etiology is identified (but prenatal diagnosis is not possible) Accompanying feature Approx. risk of recurrence Microcephaly alone 1 in 6 to 1 in 8 (10 15%) Microcephaly with other features 1 in 30 (3%) Infantile spasms 1 in 30 to 1 in 100 (1 3%)

Non specific dysmorphic features 1 in 25 to 1 in 30 (3 4%) Malformation 1 in 50 (2%) No specific features; male proband 1 in 13 (8%) No specific features; female proband 1 in 20 (5%) Holoprosencephaly with normal karyotype & no 1 in 20 (5%) forme fruste in parents Lissencephaly type 1 Very low (<1%) Lissencephaly type 2 1 in 4 (25%) Cerebellar hypoplasia 1 in 8 (12.5%) Schizencephaly/ asymmetric porencephaly Very low Cerebral palsy (diplegia/ hemiplegia) 1 in 200 to 1 in 400 Symmetrical spasticity 1 in 8 to 1 in 9 (10 12%) Asymmetric neurological signs 1 in 50 to 1 in 100 (1 2%) Ataxic diplegia 1 in 24 (4%) Congenital ataxia 1 in 8 (12.5%) Prenatal Diagnosis: Fetal karyotyping (preferably amniotic fluid): if proband has an identified chromosomal anomaly Fetal sample FISH/ MLPA/ cytogenetic microarray analysis: if proband identified to have microdeletion/ CNV detected by FISH, MLPA or microarray Fetal (CVS/ cultured amniocytes) targeted mutation analysis/ linkage analysis: if proband has monogenic disorder with identified/ unidentified mutation Fetal tissue (CVS/ cultured amniocytes/ amniotic fluid) enzyme assay/ metabolite assay: if proband identified to have a specific metabolic disorder Fetal targeted anomaly scan at 16 18 weeks and/ or serial USG monitoring for multiple malformation syndromes/ microcephaly/ CNS malformation syndromes (fetal MRI may also be done)