E-Mail Us Close
Please note that this email should only be used for feedback and comments specifically related to this particular medical policy.
  
Horizon BCBSNJ
Uniform Medical Policy ManualSection:Pathology
Policy Number:094
Effective Date: 04/14/2020
Original Policy Date:02/25/2014
Last Review Date:04/14/2020
Date Published to Web: 04/03/2017
Subject:
Whole Exome and Whole Genome Sequencing for Diagnosis of Genetic Disorders

Description:
_______________________________________________________________________________________

IMPORTANT NOTE:

The purpose of this policy is to provide general information applicable to the administration of health benefits that Horizon Blue Cross Blue Shield of New Jersey and Horizon Healthcare of New Jersey, Inc. (collectively “Horizon BCBSNJ”) insures or administers. If the member’s contract benefits differ from the medical policy, the contract prevails. Although a service, supply or procedure may be medically necessary, it may be subject to limitations and/or exclusions under a member’s benefit plan. If a service, supply or procedure is not covered and the member proceeds to obtain the service, supply or procedure, the member may be responsible for the cost. Decisions regarding treatment and treatment plans are the responsibility of the physician. This policy is not intended to direct the course of clinical care a physician provides to a member, and it does not replace a physician’s independent professional clinical judgment or duty to exercise special knowledge and skill in the treatment of Horizon BCBSNJ members. Horizon BCBSNJ is not responsible for, does not provide, and does not hold itself out as a provider of medical care. The physician remains responsible for the quality and type of health care services provided to a Horizon BCBSNJ member.

Horizon BCBSNJ medical policies do not constitute medical advice, authorization, certification, approval, explanation of benefits, offer of coverage, contract or guarantee of payment.

__________________________________________________________________________________________________________________________

Whole exome sequencing (WES) sequences the portion of the genome that contains protein-coding DNA, while whole genome sequencing (WGS) sequences both coding and noncoding regions of the genome. WES and WGS have been proposed for use in patients presenting with disorders and anomalies not explained by standard clinical workup. Potential candidates for WES and WGS include patients who present with a broad spectrum of suspected genetic conditions.

Populations
Interventions
Comparators
Outcomes
Individuals:
    • Who are children with multiple unexplained congenital anomalies or a neurodevelopmental disorder of unknown etiology following standard workup
Interventions of interest are:
    • Whole exome sequencing with trio testing when possible
Comparators of interest are:
    • Standard clinical workup without whole exome sequencing
Relevant outcomes include:
    • Test validity
    • Functional outcomes
    • Changes in reproductive decision making
    • Resource utilization
Individuals:
    • Who are children with a suspected genetic disorder other than multiple congenital anomalies or a neurodevelopmental disorder of unknown etiology following standard workup
Interventions of interest are:
    • Whole exome sequencing with trio testing when possible
Comparators of interest are:
    • Standard clinical workup without whole exome sequencing
Relevant outcomes include:
    • Test validity
    • Functional outcomes
    • Changes in reproductive decision making
    • Resource utilization
Individuals:
    • Who are children who are not critically ill with multiple unexplained congenital anomalies or a neurodevelopmental disorder of unknown etiology following standard workup
Interventions of interest are:
    • Whole genome sequencing with trio testing when possible
Comparators of interest are:
    • Standard clinical workup without whole exome sequencing
    • Whole exome sequencing with trio testing when possible
Relevant outcomes include:
    • Test validity
    • Functional outcomes
    • Changes in reproductive decision making Resource utilization
Individuals:
    • Who are children with a suspected genetic disorder other than multiple unexplained congenital anomalies or a neurodevelopmental disorder of unknown etiology following standard workup
Interventions of interest are:
    • Whole genome sequencing with trio testing when possible
Comparators of interest are:
    • Standard clinical workup without whole genome sequencing
    • Whole exome sequencing with trio testing when possible
Relevant outcomes include:
    • Test validity
    • Functional outcomes
    • Changes in reproductive decision making Resource utilization
Individuals:
    • Who are critically ill infants with a suspected genetic disorder of unknown etiology following standard workup
Interventions of interest are:
    • Rapid whole exome or genome sequencing with trio testing when possible
Comparators of interest are:
    • Standard clinical workup without whole exome or whole genome sequencing
Relevant outcomes include:
    • Test validity
    • Functional outcomes
    • Changes in reproductive decision making
    • Resource utilization

BACKGROUND

Whole Exome Sequencing and Whole Genome Sequencing

Whole exome sequencing (WES) is targeted next-generation sequencing (NGS) of the subset of the human genome that contains functionally important sequences of protein-coding DNA, while whole genome sequencing (WGS) uses NGS techniques to sequence both coding and noncoding regions of the genome. WES and WGS have been proposed for use in patients presenting with disorders and anomalies not explained by standard clinical workup. Potential candidates for WES and WGS include patients who present with a broad spectrum of suspected genetic conditions.

Given the variety of disorders and management approaches, there are a variety of potential health outcomes from a definitive diagnosis. In general, the outcomes of a molecular genetic diagnosis include (1) impacting the search for a diagnosis, (2) informing follow-up that can benefit a child by reducing morbidity, and (3) affecting reproductive planning for parents and potentially the affected patient.

The standard diagnostic workup for patients with suspected Mendelian disorders may include combinations of radiographic, electrophysiologic, biochemical, biopsy, and targeted genetic evaluations.1, The search for a diagnosis may thus become a time-consuming and expensive process.

Whole Exome Sequencing and Whole Genome Sequencing Technology

WES or WGS using NGS technology can facilitate obtaining a genetic diagnosis in patients efficiently. WES is limited to most of the protein-coding sequence of an individual (»85%), is composed of about 20000 genes and 180000 exons (protein-coding segments of a gene), and constitutes approximately 1% of the genome. It is believed that the exome contains about 85% of heritable disease-causing variants. WES has the advantage of speed and efficiency relative to Sanger sequencing of multiple genes. WES shares some limitations with Sanger sequencing. For example, it will not identify the following: intronic sequences or gene regulatory regions; chromosomal changes; large deletions; duplications; or rearrangements within genes, nucleotide repeats, or epigenetic changes. WGS uses techniques similar to WES but includes noncoding regions. WGS has a greater ability to detect large deletions or duplications in protein-coding regions compared with WES but requires greater data analytics.

Technical aspects of WES and WGS are evolving, including the development of databases such as the National Institutes of Health’s ClinVar database (http://www.ncbi.nlm.nih.gov/clinvar/) to catalog variants, uneven sequencing coverage, gaps in exon capture before sequencing, and difficulties with narrowing the large initial number of variants to manageable numbers without losing likely candidate mutations. The variability contributed by the different platforms and procedures used by different clinical laboratories offering exome sequencing as a clinical service is unknown.

In 2013, the American College of Medical Genetics and Genomics, Association for Molecular Pathology, and College of American Pathologists convened a workgroup to standardize terminology for describing sequence variants. In 2015, guidelines developed by this workgroup describe criteria for classifying pathogenic and benign sequence variants based on 5 categories of data: pathogenic, likely pathogenic, uncertain significance, likely benign, and benign.2,

Regulatory Status

Clinical laboratories may develop and validate tests in-house and market them as a laboratory service; laboratory-developed tests must meet the general regulatory standards of the Clinical Laboratory Improvement Amendments (CLIA). WES or WGS tests as a clinical service are available under the auspices of the CLIA. Laboratories that offer laboratory-developed tests must be licensed by the CLIA for high-complexity testing. To date, the U.S. Food and Drug Administration (FDA) has chosen not to require any regulatory review of this test.

Related Policies

  • Genetic Testing for Facioscapulohumeral Muscular Dystrophy (Policy #088 in the Pathology Section)
  • Genetic Testing for Epilepsy (Policy #102 in the Pathology Section)
  • Genetic Testing for Limb-Girdle Muscular Dystrophies (Policy #125 in the Pathology Section)
  • Genetic Testing for Developmental Delay/Intellectual Disability, Autism Spectrum Disorder, and Congenital Anomalies (Policy #047 in the Pathology Section)
  • Genetic Testing for the Diagnosis of Inherited Peripheral Neuropathies (Policy #087 in the Pathology Section)

Policy:
(NOTE: For services provided August 1, 2017 and after, Horizon Blue Cross Blue Shield of New Jersey collaborates with eviCore healthcare to conduct Medical Necessity Determination for certain molecular and genomic testing services for members enrolled in Horizon BCBSNJ fully insured products as well as Administrative Services Only (ASO) accounts that have elected to participate in the Molecular and Genomic Testing Program (“the Program”). Beginning August 1, 2017, the criteria and guidelines included in this policy apply to members enrolled in plans that have NOT elected to participate in the Program.

To access guidelines that apply for services provided August 1, 2017 and after to members enrolled in plans that HAVE elected to participate in the Program, please visit www.evicore.com/healthplan/Horizon_Lab.

For Medicare Advantage, Medicaid and FIDE-SNP, please refer to the Coverage Sections below for coverage guidance.)

1. Standard whole exome sequencing, with trio testing when possible (see Policy Guidelines), is considered medically necessary for the evaluation of unexplained congenital or neurodevelopmental disorder in children when ALL of the following criteria are met:

    a. Documentation that the member has been evaluated by a clinician with expertise in clinical genetics, including at minimum a family history and phenotype description, and counseled about the potential risks of genetic testing.
    b. There is potential for a change in management and clinical outcome for the individual being tested.
    c. A genetic etiology is considered the most likely explanation for the phenotype despite previous genetic testing (e.g., chromosomal microarray analysis and/or targeted single-gene testing), OR when previous genetic testing has failed to yield a diagnosis, and the affected individual is faced with invasive procedures or testing as the next diagnostic step (e.g., muscle biopsy).

2. Rapid whole exome sequencing or rapid whole genome sequencing, with trio testing when possible (see Policy Guidelines), is considered medically necessary for the evaluation of critically ill infants in neonatal or pediatric intensive care with a suspected genetic disorder of unknown etiology when BOTH of the following criteria are met:
    a. At least one of the following criteria is met:
      i. Multiple congenital anomalies (see Policy Guidelines);
      ii. An abnormal laboratory test or clinical features suggests a genetic disease or complex metabolic phenotype (see Policy Guidelines);
      iii. An abnormal response to standard therapy for a major underlying condition;
    b. None of the following criteria apply regarding the reason for admission to intensive care:
      i. An infection with normal response to therapy;
      ii. Isolated prematurity;
      iii. Isolated unconjugated hyperbilirubinemia;
      iv. Hypoxic Ischemic Encephalopathy;
      v. Confirmed genetic diagnosis explains illness;
      vi. Isolated Transient Neonatal Tachypnea;or
      vii. Nonviable neonates.

3. Whole exome sequencing is considered investigational for the diagnosis of genetic disorders in all other situations.

4. Whole genome sequencing is considered investigational for the diagnosis of genetic disorders in all other situations.

5. Whole exome sequencing and whole genome sequencing are considered investigational for screening for genetic disorders.


Medicare Coverage:
There is no National Coverage Determination (NCD) for exome sequence analysis. In the absence of an NCD, coverage decisions are left to the discretion of Local Medicare Carriers. Novitas Solutions, Inc, the Local Medicare Carrier for jurisdiction JL, has determined that CPT codes 81415, 81416, 81417, 81425, 81426 and 81427 are covered when LCD L35062 and Article A56541 criteria is met. For additional information, refer to Local Coverage Determination (LCD): Biomarkers Overview (L35062) and Local Coverage Article: Billing and Coding: Biomarkers Overview (A56541). For additional information, refer to Novitas Solutions Inc, LCD Biomarkers Overview (L35062) and Local Coverage Article: Billing and Coding: Biomarkers Overview (A56541). Available to be accessed at Novitas Solutions, Inc., Medical Policy Search page: https://www.novitas-solutions.com/webcenter/portal/MedicareJL/LcdSearch?_afrLoop=90769712476969#!%40%40%3F_afrLoop%3D90769712476969%26centerWidth%3D100%2525%26leftWidth%3D0%2525%26rightWidth%3D0%2525%26showFooter%3Dfalse%26showHeader%3Dfalse%26_adf.ctrl-state%3D63y7eftob_46.

Next generation sequencing is covered when NCD 90.2 criteria is met. For additional information and eligibility, refer to National Coverage Determination (NCD) for Next Generation Sequencing (NGS) (90.2). Available to be accessed at CMS National Coverage Determinations (NCDs) Alphabetical Index search page: https://www.cms.gov/medicare-coverage-database/indexes/ncd-alphabetical-index.aspx.

PROPRIETARY LABS (Labs that are the sole source for the diagnostic lab test)
For labs which are proprietary (that is, the sole source for the diagnostic lab test involved), Medicare Advantage Products will follow the Medicare Local Coverage Determination of the State where the proprietary lab is located.

Medicaid Coverage:

For members enrolled in Medicaid and NJ FamilyCare plans, Horizon BCBSNJ applies the above medical policy.

FIDE SNP:

For members enrolled in a Fully Integrated Dual Eligible Special Needs Plan (FIDE-SNP): (1) to the extent the service is covered under the Medicare portion of the member’s benefit package, the above Medicare Coverage statement applies; and (2) to the extent the service is not covered under the Medicare portion of the member’s benefit package, the above Medicaid Coverage statement applies.


Policy Guidelines: (Information to guide medical necessity determination based on the criteria contained within the policy statements above.)

The policy statements are intended to address the use of whole exome and whole genome sequencing for the diagnosis of genetic disorders in patients with suspected genetic disorders and for population-based screening.

This policy does not address the use of whole exome and whole genome sequencing for preimplantation genetic diagnosis or screening, prenatal (fetal) testing, or testing of cancer cells.

Rapid Sequencing

In the NSIGHT1 trial (Petrikin, 2018) rapid Whole Genome Sequencing (rWGS) provided time to provisional diagnosis by 10 days with time to final report of approximately ~ 17 days although the trial required confirmatory testing of WGS results which lengthened the time to rWGS diagnosis by 7–10 days. The WGS was performed in ‘rapid run’ mode with minimum depth of 90 Gb per genome and average depth of coverage of 40X.

For rapid WES or WGS, the patient should be critically ill and in the NICU or PICU when the test is ordered but may be discharged before results are delivered.

Copy number variation (CNV) analysis should be performed in parallel with rWGS using chromosomal microarray analysis (CMA) or directly within rWGS if the test is validated for CNV analysis.

Examples of specific malformations highly suggestive of a genetic etiology, include but are not limited to any of the following :

    • Choanal atresia
    • Coloboma
    • Hirschsprung disease
    • Meconium ileus
Examples of an abnormal laboratory test suggesting a genetic disease or complex metabolic phenotype, include but are not limited to any of the following:
    • Abnormal newborn screen
    • Conjugated hyperbilirubinemia not due to total parental nutrition (TPN) cholestasis
    • Hyperammonemia
    • Lactic acidosis not due to poor perfusion
    • Refractory or severe hypoglycemia
Examples of clinical features suggesting a genetic disease include but not limited to any of the following:
    • Significant hypotonia; or
    • Persistent seizures.
    • Infant with high risk stratification on evaluation for a Brief Resolved Unexplained Event (BRUE) (see below) with any of the following features:
        • Recurrent events without respiratory infection
        • Recurrent witnessed seizure like events
        • Required Cardiopulmonary Resuscitation (CPR)
        • Significantly abnormal chemistry including but not limited to electrolytes, bicarbonate or lactic acid, venous blood gas, glucose, or other tests that suggest an inborn error of metabolism
    • Significantly abnormal electrocardiogram (ECG), including but not limited to possible channelopathies, arrhythmias, cardiomyopathies, myocarditis or structural heart disease
    • Family history of:
        • Arrhythmia
        • BRUE in sibling
        • Developmental delay
        • Inborn error of metabolism or genetic disease
        • Long QT syndrome (LQTS)
        • Sudden unexplained death (including unexplained car accident or drowning) in first- or second-degree family members before age 35, and particularly as an infant
BRUE

Brief Resolved Unexplained Event (BRUE) was previously known as Apparent Life Threatening Event (ALTE). In a practice guideline from the American Academy of Pediatrics (AAP), BRUE is defined as an event occurring in an infant younger than 1 year of age when the observer reports a sudden, brief (usually less than one minute), and now resolved episode of one or more of the following:

    • Absent, decreased, or irregular breathing
    • Altered level of responsiveness
    • Cyanosis or pallor
    • Marked change in tone (hyper- or hypotonia)
A BRUE is diagnosed only when there is no explanation for a qualifying event after conducting an appropriate history and physical examination.
Note: More information is available at:
https://pediatrics.aappublications.org/content/137/5/e20160590

Trio Testing

The recommended option for testing when possible is testing of the child and both parents (trio testing). Trio testing increases the chance of finding a definitive diagnosis and reduces false-positive findings.

Trio testing is preferred whenever possible but should not delay testing of a critically ill patient when rapid testing is indicated. Testing of one available parent should be done if both are not immediately available and one or both parents can be done later if needed.

Genetics Nomenclature Update

The Human Genome Variation Society nomenclature is used to report information on variants found in DNA and serves as an international standard in DNA diagnostics. It is being implemented for genetic testing medical policy updates starting in 2017 (see Table PG1). The Society’s nomenclature is recommended by the Human Variome Project, the HUman Genome Organization, and by the Human Genome Variation Society itself.

The American College of Medical Genetics and Genomics and the Association for Molecular Pathology standards and guidelines for interpretation of sequence variants represent expert opinion from both organizations, in addition to the College of American Pathologists. These recommendations primarily apply to genetic tests used in clinical laboratories, including genotyping, single genes, panels, exomes, and genomes. Table PG2 shows the recommended standard terminology¾“pathogenic,” “likely pathogenic,” “uncertain significance,” “likely benign,” and “benign”¾to describe variants identified that cause Mendelian disorders.

Table PG1. Nomenclature to Report on Variants Found in DNA
PreviousUpdatedDefinition
MutationDisease-associated variantDisease-associated change in the DNA sequence
 VariantChange in the DNA sequence
 Familial variantDisease-associated variant identified in a proband for use in subsequent targeted genetic testing in first-degree relatives

Table PG2. ACMG-AMP Standards and Guidelines for Variant Classification
Variant ClassificationDefinition
Likely pathogenicLikely disease-causing change in the DNA sequence
PathogenicDisease-causing change in the DNA sequence
Variant of uncertain significanceChange in DNA sequence with uncertain effects on disease
Likely benignLikely benign change in the DNA sequence
BenignBenign change in the DNA sequence
ACMG: American College of Medical Genetics and Genomics; AMP: Association for Molecular Pathology.

Genetic Counseling

Experts recommend formal genetic counseling for patients who are at risk for inherited disorders and who wish to undergo genetic testing. Interpreting the results of genetic tests and understanding risk factors can be difficult for some patients; genetic counseling helps individuals understand the impact of genetic testing, including the possible effects the test results could have on the individual or their family members. It should be noted that genetic counseling may alter the utilization of genetic testing substantially and may reduce inappropriate testing; further, genetic counseling should be performed by an individual with experience and expertise in genetic medicine and genetic testing methods.


[RATIONALE: The policy was created in 2014 and has been updated regularly with searches of the MEDLINE database. The most recent literature update was performed through January 31, 2020. Evidence reviews assess whether a medical test is clinically useful. A useful test provides information to make a clinical management decision that improves the net health outcome. That is, the balance of benefits and harms is better when the test is used to manage the condition than when another test or no test is used to manage the condition.

The first step in assessing a medical test is to formulate the clinical context and purpose of the test. The test must be technically reliable, clinically valid, and clinically useful for that purpose. Evidence reviews assess the evidence on whether a test is clinically valid and clinically useful. Technical reliability is outside the scope of these reviews, and credible information on technical reliability is available from other sources.

Overview

This review was informed in part by a TEC Special Report (2013) on exome sequencing for patients with suspected genetic disorders.3,

In 2018, Smith et al reported a scoping review of genome and exome sequencing as a diagnostic tool for pediatric patients. 4, The authors identified 171 publications) although 131 were case reports. They concluded that diagnostic yield was the only consistently reported outcome. The median diagnostic yield in publications including more than single case reports was 33% but varied by broad clinical categories and test type.

The following sections review evidence by test type (WES and WGS), broad type of disorder and care setting (intensive care vs. not intensive care).

Whole Exome Sequencing for Children with Multiple Congenital Anomalies or a Neurodevelopmental Disorder of Unknown Etiology Following Standard Workup; Patients who are not Critically Ill
Clinical Context and Test Purpose

The purpose of whole exome sequencing (WES) in children who have multiple unexplained congenital anomalies or a neurodevelopmental disorder of unknown etiology following standard workup is to establish a molecular diagnosis. The criteria under which diagnostic testing for a genetic or heritable disorder may be considered clinically useful are as follows:

    • A definitive diagnosis cannot be made based on history, physical examination, pedigree analysis, and/or standard diagnostic studies or tests;
    • The clinical utility of a diagnosis has been established (eg, by demonstrating that a definitive diagnosis will lead to changes in clinical management of the condition, changes in surveillance, or changes in reproductive decision making, and these changes will lead to improved health outcomes); and
    • Establishing the diagnosis by genetic testing will end the clinical workup for other disorders.
The question addressed in this policy is: Does the use of WES improve health outcomes when used for the diagnosis of children with multiple unexplained congenital anomalies or a neurodevelopmental disorder of unknown etiology following standard workup?

The following PICO was used to select literature to inform this review.

Patients

The relevant population of interest is children presenting with multiple unexplained congenital anomalies or a neurodevelopmental disorder that are suspected to have a genetic basis but are not explained by standard clinical workup.

Intervention

The relevant intervention of interest is WES with trio testing when possible.

Comparators

The following practice is currently being used to diagnose multiple unexplained congenital anomalies or a neurodevelopmental disorder: standard clinical workup without WES.

A standard clinical workup for an individual with a suspected genetic condition varies by patient phenotype but generally involves a thorough history, physical exam (including dysmorphology and neurodevelopmental assessment, if applicable), routine laboratory testing, and imaging. If the results suggest a specific genetic syndrome, then established diagnostic methods relevant for that syndrome would be used.

Outcomes

There is no reference standard for the diagnosis of patients who have exhausted alternative testing strategies, therefore diagnostic yield will be the clinical validity outcome of interest.

The health outcomes of interest are reduction in morbidity due to appropriate treatment and surveillance, the end of the diagnostic odyssey, and effects on reproductive planning for parents and potentially the affected patient.

False-positive test results can lead to misdiagnosis and inappropriate clinical management. False-negative test results can lead to a lack of a genetic diagnosis and continuation of the diagnostic odyssey.

Study Selection Criteria

For the evaluation of clinical validity of WES, studies that met the following eligibility criteria were considered:

    • Reported on the diagnostic yield or performance characteristics such as sensitivity and specificity of WES;
    • Patient/sample clinical characteristics were described; children with congenital abnormalities or neurodevelopmental disorders were included;
    • Patient/sample selection criteria were described;
    • Included at least 20 patients.
Technically Reliable

Assessment of technical reliability focuses on specific tests and operators and requires review of unpublished and often proprietary information. Review of specific tests, operators, and unpublished data are outside the scope of this policy and alternative sources exist. This policy focuses on the clinical validity and clinical utility.

Clinically Valid

A test must detect the presence or absence of a condition, the risk of developing a condition in the future, or treatment response (beneficial or adverse).

A number of studies have reported on the use of WES in clinical practice (see Table 1). Typically, the populations included in these studies have had suspected rare genetic disorders, although the specific populations vary.

Series have been reported with as many as 2000 patients. The most common reason for referral to a tertiary care center was an unexplained neurodevelopmental disorder. Many patients had been through standard clinical workup and testing without identification of a genetic variant to explain their condition. Diagnostic yield in these studies, defined as the proportion of tested patients with clinically relevant genomic abnormalities, ranged from 25% to 48%. Because there is no reference standard for the diagnosis of patients who have exhausted alternative testing strategies, clinical confirmation may be the only method for determining false-positive and false-negative rates. No reports were identified of incorrect diagnoses, and how often they might occur is unclear.

When used as a first-line test in infants with multiple congenital abnormalities and dysmorphic features, diagnostic yield may be as high as 58%. Testing parent-child trios has been reported to increase diagnostic yield, to identify an inherited variant from an unaffected parent and be considered benign, or to identify a de novo variant not present in an unaffected parent. First-line trio testing for children with complex neurologic disorders was shown to increase the diagnostic yield (29%, plus a possible diagnostic finding in 27%) compared with a standard clinical pathway (7%) performed in parallel in the same patients.5,

Table 1. Diagnostic Yields of Whole Exome Sequencing for Congenital Anomalies or a Neurodevelopmental Disorder
Study
Patient Population
N
Design
Yield, n (%)
Additional Information
Cordoba et al (2018)6,Patients suspected of having a neurogenetic condition: typical findings of known neurogenetic diseases and/or
hints of monogenic etiology such as familial aggregation or chronic and progressive courseMean age was 23 yrs
40Prospective
Consecutive patients selected from a Neurogenetic Clinic of a tertiary Hospital in ArgentinaUnclear how many were trio testing
16 (40)Results led to altered treatment in 14 patients
Ewans et al (2018)7,Patients from families with a distinctive phenotype likely to
have a monogenic etiology with a family structure consistent with Mendelian inheritance. Prior diagnostic testing had all been negative.. The majority of disorders were intellectual disability or neurological (62%) but 13% were skeletal and 11% were hematologicalTwo-thirds pediatric
37 families54 individuals from 37 families recruited from clinical genetics units in NewSouth Wales from 2013 to 2014. Proband plus family members(s) underwent WES.11 (30)Reanalysis at 12 mos improved diagnostic success from 30 to 41%
Powis et al (2018) 8,Neonates (birth to 1 mo of age). The majority had multiple congenital anomalies or dysmorphic features.66Trio or singleton WES6 infants received rapid WESOverall: 25 (38)rapid WES: 3 (50)VUS noted in 6 patients
Wright et al (2018)9,, re-analysisWright et al (2015)10,, original analysisChildren with severe undiagnosed NDDs and/or congenital anomalies, abnormal growth parameters, dysmorphic features, and unusual behavioral phenotypes1133Consecutive family trios from U.K.-wide patient recruitment network454 (40), re-analysis
311 (27), original analysis
Wright et al (2018) is reanalysis of existing data from earlier Wright et al (2015) publication from DDD study using improved variant calling methodologies, novel variant detection algorithms, updated variant annotation, evidence-based filtering strategies, and newly discovered disease-associated genes
Nambot et al (2018)11,Children with congenital anomalies and intellectual disability with negative prior diagnostic workup461Consecutive cases meeting criteria referred to specialty clinic in France31%Initial yield in y 1: 22%, reanalysis led to increase yield
Tsuchida et al (2018)12,Children with epilepsy (»63% with early-onset epileptic encephalopathies) with no causative SNV in known epilepsy-associated genes168Consecutive unsolved cases referred to a single-center18 (11)Performed WES with CNV detection tools
Evers et al (2017)13,Children with undiagnosed NDDs (63%), neurometabolic disorders, and dystonias72Prospective study, referral and selection unclear
    • 36% in NDD
    • 43% in neurometabolic disorders
    • 25% in dystonias
Results reported to be important for family planning, used for a prenatal diagnostic procedure in 4 cases, management changes reported in 8 cases; surveillance for other disease-associated complications initiated in 6 cases
Vissers et al (2017)5,Children with complex neurologic disorders of suspected genetic origin150Prospective comparative study at a tertiary center
    • 44 (29) conclusive
    • 41 (27) possible
First-line WES had 29% yield vs. 7% yield for standard diagnostic workupb
Nolan and Carlson (2016)14,Children with unexplained NDDs50Pediatric neurology clinic41 (48)Changed medication, systemic investigation, and family planning
Allen et al (2016)15,Patients with unexplained early-onset epileptic encephalopathy50 (95% <1 y)Single-center11 (22)2 VUS for follow-up, 11 variants identified as de novo
Stark et al (2016)16,Infants (≤2 y) with suspected monogenic disorders with multiple congenital abnormalities and dysmorphic features80 overall;37 critically illProspective comparative study at a tertiary center46 (58) overall;19 (51) in critically ill infantsFirst-line WES increased yield by 44%, changed clinical management and family planning.
Tarailo-Graovac et al (2016)17,Intellectual developmental disorders and unexplained metabolic phenotypes (all ages)41Consecutively enrolled patients referred to a single-center28 (68)WES diagnosis affected the clinical treatment of 18 (44%) probands
Farwell et al (2015)18,Unexplained neurologic disorders (65% pediatric)500WES laboratory152 (30)Trio (37.5% yield) vs. proband only (20.6% yield); 31 (7.5% de novo)
Yang et al (2014)19,Suspected genetic disorder (88% neurologic or developmental)2000 (45% <5 y; 42% 5-18 yrs; 12% adults)Consecutive patients at single-center504 (25)Identification of novel variants. End of the diagnostic odyssey and change in management
Lee et al (2014)20,Suspected rare Mendelian disorders (57% of children had developmental delay; 26% of adults had ataxia)814 (49% <5 y; 15% 5-18 y; 36% adults)Consecutive patients at single-center213 (26)Trio (31% yield) vs. proband only (22% yield)
Iglesias et al (2014)21,Birth defects (24%); developmental delay (25%); seizures (32%)115 (79% children)Single-center tertiary clinic37 (32)Discontinuation of planned testing, changed medical management, and family planning
Soden et al (2014)22,Children with unexplained NDDs119 (100 families)Single-center databasea53 (45)Change in clinical care or impression in 49% of families
Srivastava et al (2014)23,Children with unexplained NDDs78Pediatric neurogenetics clinic32 (41)Change in medical management, prognostication, and family planning
Yang et al (2013)24,Suspected genetic disorder (80% neurologic)250 (1% fetus; 50% <5 y; 38% 5-18 yrs; 11% adults)Consecutive patients at single-center62 (25)Identification of atypical phenotypes of known genetic diseases and blended phenotypes

CNV: copy number variant; DDD: Deciphering Developmental Disorders; NDD: neurodevelopmental disorder; SNV: single nucleotide variants; VUS: variant of uncertain significance; WES: whole exome sequencing.
a Included both WES and whole genome sequencing.
b Standard diagnostic workup included an average of 23.3 physician-patient contacts, imaging studies, muscle biopsies or lumbar punctures, other laboratory tests, and an average of 5.4 sequential gene by gene tests.
Clinically Useful

A test is clinically useful if the use of the results informs management decisions that improve the net health outcome of care. The net health outcome can be improved if patients receive correct therapy, or more effective therapy, or avoid unnecessary therapy, or avoid unnecessary testing.

Direct Evidence

Direct evidence of clinical utility is provided by studies that have compared health outcomes for patients managed with and without the test. Because these are intervention studies, the preferred evidence would be from randomized controlled trials (RCTs).

No RCTs assessing the use of WES to diagnose multiple unexplained congenital anomalies or a neurodevelopmental disorder were identified.

Chain of Evidence

Indirect evidence on clinical utility rests on clinical validity. If the evidence is insufficient to demonstrate test performance, no inferences can be made about clinical utility.

Cohort studies following children from presentation to outcomes have not been reported. There are considerable challenges conducting studies of sufficient size given the underlying genetic heterogeneity, and including follow-up adequate to observe final health outcomes. Studies addressing clinical utility have reported mainly diagnostic yield and management changes. Thus, it is difficult to quantify lower or upper bounds for any potential improvement in the net health outcome owing in part to the heterogeneity of disorders, rarity, and outcome importance that may differ according to identified pathogenic variants. Actionable items following testing in the reviewed studies (see Table 2) included family planning, change in management, change or avoidance of additional testing, surveillance for associated morbidities, prognosis, and ending the diagnostic odyssey.

The evidence reviewed here reflects the accompanying uncertainty, but supports a perspective that identifying a pathogenic variant can (1) impact the search for a diagnosis, (2) inform follow-up that can benefit a child by reducing morbidity and rarely potential mortality, and (3) affect reproductive planning for parents and later potentially the affected child. When recurrence risk can be estimated for an identified variant (eg, by including parent testing), future reproductive decisions can be affected. Early use of WES can reduce the time to diagnosis and reduce the financial and psychological burdens associated with prolonged investigation.

Section Summary: Whole Exome Sequencing for Children with Multiple Congenital Anomalies or a Neurodevelopmental Disorder of Unknown Etiology Following Standard Workup

The evidence on WES in children who have multiple congenital anomalies or a developmental disorder with a suspected genetic etiology of unknown etiology following standard workup includes case series. These series have reported diagnostic yields of WES ranging from 22% to 58%, depending on the individual’s age, phenotype, and previous workup. Comparative studies have reported an increase in diagnostic yield compared with standard testing strategies. Thus, for individuals who have a suspected genetic etiology but for whom the specific genetic alteration is unclear or unidentified by standard clinical workup, WES may return a likely pathogenic variant. A genetic diagnosis for these patients is reported to change management, including medication changes, discontinuation of or additional testing, ending the diagnostic odyssey, and family planning.

Whole Exome Sequencing for Children with a Suspected Genetic Disorder other than Multiple Congenital Anomalies or a Neurodevelopmental Disorder of Unknown Etiology Following Standard Workup; Patients who are not Critically Ill
Clinical Context and Test Purpose

Most of the literature on WES is on neurodevelopmental disorders in children; however, other potential indications for WES have been reported (see Table 3). These include limb-girdle muscular dystrophy, inherited retinal disease, and other disorders including mitochondrial, endocrine, and immunologic disorders.

The purpose of WES in patients who have a suspected genetic disorder other than multiple unexplained congenital anomalies or a neurodevelopmental disorder of unknown etiology following standard workup is to establish a molecular diagnosis. The criteria under which diagnostic testing for a genetic or heritable disorder may be considered clinically useful are stated above.

The question addressed in this policy is: Does WES improve health outcomes when used for the diagnosis of a suspected genetic condition other than multiple congenital anomalies or a neurodevelopmental disorder of unknown etiology following standard workup?

The following PICO was used to select literature to inform this review.

Patients

The relevant population of interest is children presenting with a disorder other than multiple unexplained congenital anomalies or a neurodevelopmental disorder that is suspected to have a genetic basis but is not explained by standard clinical workup.

Intervention

The relevant intervention of interest is WES. Specific tests were described in the preceding section on WES.

Comparators

The following practice is currently being used to diagnose a suspected genetic disorder other than multiple unexplained congenital anomalies or a neurodevelopmental disorder: standard clinical workup without WES.

Standard clinical workup was described in a preceding section.

Outcomes

There is no reference standard for the diagnosis of patients who have exhausted alternative testing strategies, therefore diagnostic yield will be the clinical validity outcome of interest.

The health outcomes of interest are reduction in morbidity due to appropriate treatment and surveillance, the end of the diagnostic odyssey, and effects on reproductive planning for parents and potentially the affected patient.

Study Selection Criteria

For the evaluation of clinical validity of WES, studies that met the following eligibility criteria were considered:

    • Reported on the diagnostic yield or performance characteristics such as sensitivity and specificity of WES;
    • Patient/sample clinical characteristics were described;
    • Patient/sample selection criteria were described;
    • Included at least 20 patients.
Technically Reliable

Assessment of technical reliability focuses on specific tests and operators and requires review of unpublished and often proprietary information. Review of specific tests, operators, and unpublished data are outside the scope of this policy and alternative sources exist. This policy focuses on the clinical validity and clinical utility.

Clinically Valid

A test must detect the presence or absence of a condition, the risk of developing a condition in the future, or treatment response (beneficial or adverse).

Studies have assessed WES for a broad spectrum of disorders. The diagnostic yield in patient populations restricted to specific phenotypes ranges from 3% for colorectal cancer to 60% for unexplained limb-girdle muscular dystrophy (see Table 2). Some studies used a virtual gene panel that is restricted to genes associated with the phenotype, while others have examined the whole exome, either initially or sequentially. An advantage of WES over individual gene or gene panel testing is that the stored data allows reanalysis as new genes are linked to the patient phenotype. WES has also been reported to be beneficial in patients with atypical presentations.

Table 2. Diagnostic Yields of Whole Exome Sequencing for Conditions Other Than Multiple Congenital Anomalies or a Neurodevelopmental Disorder
Study
Patient Population
N
Design
Yield,n (%)
Additional Information
Hauer et al (2018)25,Short stature in whom common nongenetic causes had been excluded200 (mostly children)Randomly selected from a consecutive series of patients referred for workup; trio testing performed33 (17)
    • Standard diagnostic approach yield: 13.6% in original cohort of 565
    • WES results had possible impact on treatment or additional preventive measurements in 31 (16%) families
Rossi et al (2017)26,Patients with autism spectrum disorder diagnosis or autistic features referred for WES163Selected from 1200 consecutive retrospective samples from commercial lab42 (26)
    • 66% of patients already had a clinician-reported autism diagnosis
    • VUS in 12%
Walsh et al (2017)27,Peripheral neuropathy in patients ranging from 2-68 y
    • 23 children
    • 27 adults
Prospective research study at tertiary pediatric and adult centers19 (38)Initial targeted analysis with virtual gene panel, followed by WES
Miller et al (2017)28,Craniosynostosis in patients who tested negative on targeted genetic testing40Research study of referred patientsa15 (38)Altered management and reproductive decision making
Posey et al (2016)29,Adults (overlap of 272 patients reported by Yang et al [2014]),19, includes neurodevelopmental and other phenotypes486 (53% 18-30 y; 47% >30 y)Review of lab findings in consecutive retrospective series of adults85 (18)Yield in patients 18-30 y (24%) vs. those >30 y (10.4%)
Ghaoui et al (2015)30,Unexplained limb-girdle muscular dystrophy60 familiesProspective study of patients identified from specimen bank27 (60)Trio (60% yield) vs. proband only (40% yield)
Valencia et al (2015)31,Unexplained disorders: congenital anomalies (30%), neurologic (22%), mitochondrial (25%), endocrine (3%), immunodeficiencies (17%)40 (<17 y)Consecutive patients in a single-center12 (30)
    • Altered management including genetic counseling and ending diagnostic odyssey
    • VUS in 15 (38%) patients
Wortmann et al (2015)32,Suspected mitochondrial disorder109Patients referred to a single-center42 (39)57% yield in patients with high suspicion of mitochondrial disorder
Neveling et al (2013)33,Unexplained disorders: blindness, deafness, movement disorders, mitochondrial disorders, hereditary cancer186Outpatient genetic clinic; post hoc comparison with Sanger sequencing3%-52%WES increased yield vs. Sanger sequencing Highest yield for blindness and deafness
WES: whole exome sequencing; VUS: variant of uncertain significance.
a
 Included both WES and whole genome sequencing.

The purpose of the limitations tables (see Tables 3 and 4) is to display notable limitations identified in each study. This information is synthesized as a summary of the body of evidence and provides the conclusions on the sufficiency of the evidence supporting the position statement.

Table 3. Relevance Limitations for Studies Assessing Whole Exome Sequencing for Conditions Other Than Multiple Congenital Anomalies or a Neurodevelopmental Disorder
Study
Populationa
Interventionb
Comparatorc
Outcomesd
Duration of Follow-Upe
Hauer et al (2018)25,
Rossi et al (2017)26,4. Most patients had a clinical diagnosis; only 33% had testing for specific ASD genes before WES
Walsh et al (2017)27,3. Proband testing only
Miller et al (2017)28,
Posey et al (2016)29,3. Included highly heterogeneous diseases3. Proband testing only
Ghaoui et al (2015)30,
Valencia et al (2015)31,3. Included highly heterogeneous diseases2. Unclear whether WES performed on parents
Wortmann et al (2015)32,3. Proband testing only
Neveling et al (2013)33,3. Included highly heterogeneous diseases3. Proband testing only
The study limitations stated in this table are those notable in the current review; this is not a comprehensive limitations assessment.
ASD: autism spectrum disorder; WES: whole exome sequencing.

Population key: 1. Intended use population unclear; 2. Clinical context is unclear; 3. Study population is unclear; 4. Study population not representative of intended use.

Intervention key: 1. Classification thresholds not defined; 2. Version used unclear; 3. Not intervention of interest.

c Comparator key: 1. Classification thresholds not defined; 2. Not compared to credible reference standard; 3. Not compared to other tests in use for same purpose.

d Outcomes key: 1. Study does not directly assess a key health outcome; 2. Evidence chain or decision model not explicated; 3. Key clinical validity outcomes not reported (sensitivity, specificity, and predictive values); 4. Reclassification of diagnostic or risk categories not reported; 5. Adverse events of the test not described (excluding minor discomforts and inconvenience of venipuncture or noninvasive tests).

e Follow-Up key: 1. Follow-up duration not sufficient with respect to natural history of disease (true-positives, true-negatives, false-positives, false-negatives cannot be determined).

Table 4. Study Design and Conduct Limitations for Studies Assessing Whole Exome Sequencing for Conditions Other Than Multiple Congenital Anomalies or a Neurodevelopmental Disorder
Study
Selectiona
Blindingb
Delivery of Testc
Selective Reportingd
Data Completenesse
Statisticalf
Hauer et al (2018)25,
Rossi et al (2017)26,
Walsh et al (2017)27,
Miller et al (2017)28,2. Selection not random or consecutive
Posey et al (2016)29,
Ghaoui et al (2015)30,
Valencia et al (2015)31,
Wortmann et al (2015)32,1,2. Unclear how patients were selected from those eligible
Neveling et al (2013)33,1,2. Unclear how patients were selected from those referred
The study limitations stated in this table are those notable in the current review; this is not a comprehensive limitations assessment.
WES: whole exome sequencing.

Selection key: 1. Selection not described; 2. Selection not random or consecutive (ie, convenience).

Blinding key: 1. Not blinded to results of reference or other comparator tests.

cTest Delivery key: 1. Timing of delivery of index or reference test not described; 2. Timing of index and comparator tests not same; 3. Procedure for interpreting tests not described; 4. Expertise of evaluators not described.

d Selective Reporting key: 1. Not registered; 2. Evidence of selective reporting; 3. Evidence of selective publication.

e Data Completeness key: 1. Inadequate description of indeterminate and missing samples; 2. High number of samples excluded; 3. High loss to follow-up or missing data.

f Statistical key: 1. Confidence intervals and/or p values not reported; 2. Comparison with other tests not reported.

Clinically Useful

A test is clinically useful if the use of the results informs management decisions that improve the net health outcome of care. The net health outcome can be improved if patients receive correct therapy, or more effective therapy, or avoid unnecessary therapy, or avoid unnecessary testing.

Direct Evidence

Direct evidence of clinical utility is provided by studies that have compared health outcomes for patients managed with and without the test. Because these are intervention studies, the preferred evidence would be from RCTS.

No RCTs assessing the use of WES to diagnose a suspected genetic disorder other than multiple unexplained congenital anomalies or a neurodevelopmental disorder were identified.

Chain of Evidence

Indirect evidence on clinical utility rests on clinical validity. If the evidence is insufficient to demonstrate test performance, no inferences can be made about clinical utility.

A genetic diagnosis for an unexplained disorder can alter management in several ways: such a diagnosis may lead to including genetic counseling and ending the diagnostic odyssey and may affect reproductive decision making.

Because the clinical validity of WES for this indication has not been established, a chain of evidence cannot be constructed.

Section Summary: WES for a Suspected Genetic Disorder Other Than Multiple Congenital Anomalies or a Neurodevelopmental Disorder

There is an increasing number of reports assessing use of WES identify a molecular basis for disorders other than multiple congenital anomalies or neurodevelopmental disorders. The diagnostic yields in these studies ranged from 3% for colorectal cancer to 60% for trio (parents and child) analysis of limb-girdle muscular dystrophy. Some studies have reported on the use of a virtual gene panel with restricted analysis of disease-associated genes, and the authors noted that WES data allows reanalysis as new genes are linked to the patient phenotype. Overall, a limited number of patients have been studied for any specific disorder, and study of WES in these disorders is at an early stage with uncertainty about changes in patient management.

Whole Genome Sequencing for Children with Multiple Congenital Anomalies or a Neurodevelopmental Disorder of Unknown Etiology Following Standard Workup; Patients who are not Critically Ill

The purpose of whole genome sequencing (WGS) in patients with a suspected genetic disorder of unknown etiology following standard workup is to establish a molecular diagnosis from either the coding or noncoding regions of the genome. The criteria under which diagnostic testing for a genetic or heritable disorder may be considered clinically useful are stated above.

The question addressed in this policy is: Does WGS improve health outcomes when used for the diagnosis of patients with a suspected genetic disorder of unknown etiology following standard workup without WES or WGS?

The following PICO was used to select literature to inform this review.

Patients

The relevant population of interest is:

    • Children who are not critically ill with multiple unexplained congenital anomalies or a neurodevelopmental disorder of unknown etiology following standard workup.
Interventions

The relevant interventions being considered include:

    • WGS with trio testing when possible
Several laboratories offer WGS as a clinical service. Medical centers may also offer r WGS as a clinical service.

The median time for standard WGS is several weeks.

Note that this policy does not address the use of WGS for preimplantation genetic diagnosis or screening, prenatal (fetal) testing, or for testing of cancer cells.

Comparators

The following practice is currently being used to diagnose a suspected genetic disorder: standard clinical workup without WES or WGS.

Standard clinical workup was described in a preceding section.

Outcomes

Outcomes of interest are as described above for use of WES in patients with multiple congenital anomalies or a neurodevelopmental disorder.

Study Selection Criteria

For the evaluation of clinical validity of WGS, studies that met the following eligibility criteria were considered:

    • Reported on the diagnostic yield or performance characteristics such as sensitivity and specificity of rapid WGS or WGS;
    • Patient/sample clinical characteristics were described;
    • Patient/sample selection criteria were described;
    • Included at least 20 patients.
Technically Reliable

Assessment of technical reliability focuses on specific tests and operators and requires review of unpublished and often proprietary information. Review of specific tests, operators, and unpublished data are outside the scope of this policy and alternative sources exist. This policy focuses on the clinical validity and clinical utility.

Clinically Valid

A test must detect the presence or absence of a condition, the risk of developing a condition in the future, or treatment response (beneficial or adverse).

Studies have shown that WGS can detect more pathogenic variants than WES, due to an improvement in detecting copy number variants, insertions and deletions, intronic single-nucleotide variants, and exonic single-nucleotide variants in regions with poor coverage on WES. A majority of studies described methods for interpretation of WGS indicating that only pathogenic or likely pathogenic variants were included in the diagnostic yield and that VUS were not reported. In some studies, the genes examined were those previously associated with the phenotype, while other studies were research-based and conducted more exploratory analysis.34, It has been noted that genomes sequenced with WGS are available for future review when new variants associated with clinical diseases are discovered.

The use of WGS has been studied in children who are not critically ill with multiple unexplained congenital anomalies or a neurodevelopmental disorder of unknown etiology following standard workup in several observational studies, both prospective and retrospective. Studies are described in Table 5. The diagnostic yield of WGS has been between 20% and 40%. Additional indirect evidence is available from studies reporting diagnostic yield of WES in a similar population as summarized above, and it is reasonable to expect that WGS is likely to result in similar or better diagnostic yield for pathogenic or likely pathogenic variants as compared with WES.

Table 5. Diagnostic Yields with Whole Genome Sequencing in Children who are not Critically Ill with Multiple Unexplained Congenital Anomalies or a Neurodevelopmental Disorder of Unknown Etiology Following Standard Workup
Study
Patient Population
N
Design
Yield,n (%)
Additional Information
Lionel et al (2018)34,Well-characterized but genetically heterogeneous cohort of children <18 y that had undergone targeted gene sequencing
Referral clinic: 44% metabolic, 23% ophthalmology, 15% Joint laxity/hypermobility
103ProspectiveTrio WGS testing for patients recruited from pediatric nongenetic subspecialists42 (41)Compared with a 24% yield with standard diagnostic testing and a 25% increase in yield from WES
Limited information on change in management
Costain et al (2018), re-analysis35,
Stavropoulos et al (2016)
36,, original analysis
Children (<18 y) with an undiagnosed congenital malformations and neurodevelopmental disorders
Presentation: abnormalities of the nervous system (77%), skeletal system (68%), growth (44%), eye (34%), cardiovascular (32%) and musculature (27%)
64, re-analysis
100, original analysis
Prospective, consecutive
Proband WGS was offered in parallel with clinicalCMA testing
7 (11), re-analysis

34 (34), original analysis
Costain (2018) is re-analysis of undiagnosed patients from Stavropoulos et al (2016)
CMA plus targeted gene sequencing yield was 13%WGS yield highest for developmental delay 39% (22/57) and lowest (15%) for connective tissue disorders
Change in management reported for some patients
7 incidental findings
Hiatt et al (2018) 37, re-analysisBowling et al (2017)38, original analysisChildren with developmental and/or intellectual delays of unknown etiology
81% had genetic testing prior to enrollment
Original analysis included 244
Re-analysis includes additional 123, for a total cohort of 494
Retrospective, selection method and criteria unclear
Trio WGS in a referral center
54 (22)1, original analysisRe-analysis:Re-analysis yielded pathogenic or likely pathogenic variants that were not initially reported in 23 patients
Downgraded 3 'likely pathogenic' and 6 VUS
Original analysis:Compared to 30% yield for WES
1
Changes in management not reported
11% VUS in WGS
Gilissen et al (2014)39,Children with severe intellectual disability who did not have a diagnosis after extensive genetic testing that included whole exome sequencing50Trio WGS testing including unaffected parents201 (42)Of 21 with positive diagnosis, 20 had de novo variants
Changes in management not reported
VUS: variant of uncertain significance; WGS: whole genome sequencing; WES: whole exome sequencing; CMA: chromosomal microarray analysis.

1 SNV/indel

Tables 6 and 7 display notable limitations identified in each study.

Table 6. Relevance Limitations for Studies of Whole Genome Sequencing
Study
Populationa
Interventionb
Comparatorc
Outcomesd
Duration of Follow-Upe
Lionel et al (2018)34,3. Included highly heterogeneous diseases3. Proband testing only
Costain et al (2018), re-analysis35,3. Proband testing only
Bowling et al (2017)38,4. 19% had no prescreening performed
Gilissen et al (2014)39,
The study limitations stated in this table are those notable in the current review; this is not a comprehensive limitations assessment.
WGS: whole genome sequencing.

Population key: 1. Intended use population unclear; 2. Clinical context is unclear; 3. Study population is unclear; 4. Study population not representative of intended use.
b
Intervention key: 1. Classification thresholds not defined; 2. Version used unclear; 3. Not intervention of interest.
c
 Comparator key: 1. Classification thresholds not defined; 2. Not compared to credible reference standard; 3. Not compared to other tests in use for same purpose.
d
 Outcomes key: 1. Study does not directly assess a key health outcome; 2. Evidence chain or decision model not explicated; 3. Key clinical validity outcomes not reported (sensitivity, specificity, and predictive values); 4. Reclassification of diagnostic or risk categories not reported; 5. Adverse events of the test not described (excluding minor discomforts and inconvenience of venipuncture or noninvasive tests).
e
 Follow-Up key: 1. Follow-up duration not sufficient with respect to natural history of disease (true-positives, true-negatives, false-positives, false-negatives cannot be determined).

Table 7. Study Design and Conduct Limitations for Studies of Whole Genome Sequencing
Study
Selectiona
Blindingb
Delivery of Testc
Selective Reportingd
Data Completenesse
Statisticalf
Lionel et al (2018)34,1,2. Unclear how patients were selected from those eligible
Costain et al (2018), re-analysis35,
Bowling et al (2017)38,1,2. Unclear how patients were selected from those eligible
Gilissen et al (2014)39,
The study limitations stated in this table are those notable in the current review; this is not a comprehensive limitations assessment. WGS: whole genome sequencing.
Selection key: 1. Selection not described; 2. Selection not random or consecutive (ie, convenience).
Blinding key: 1. Not blinded to results of reference or other comparator tests.
Test Delivery key: 1. Timing of delivery of index or reference test not described; 2. Timing of index and comparator tests not same; 3. Procedure for interpreting tests not described; 4. Expertise of evaluators not described.
d
 Selective Reporting key: 1. Not registered; 2. Evidence of selective reporting; 3. Evidence of selective publication.
e
 Data Completeness key: 1. Inadequate description of indeterminate and missing samples; 2. High number of samples excluded; 3. High loss to follow-up or missing data.
f
 Statistical key: 1. Confidence intervals and/or p values not reported; 2. Comparison with other tests not reported.

Clinically Useful

A test is clinically useful if the use of the results informs management decisions that improve the net health outcome of care. The net health outcome can be improved if patients receive correct therapy, or more effective therapy, or avoid unnecessary therapy, or avoid unnecessary testing.

Direct Evidence

Direct evidence of clinical utility is provided by studies that have compared health outcomes for patients managed with and without the test. Because these are intervention studies, the preferred evidence would be from RCTs.

No RCTs assessing the use of WGS to diagnose multiple unexplained congenital anomalies or a neurodevelopmental disorder outside of critical care were identified.

Chain of Evidence

Indirect evidence on clinical utility rests on clinical validity. If the evidence is insufficient to demonstrate test performance, no inferences can be made about clinical utility.

Clinical validity is established based on the meaningful diagnostic yield associated with WGS when a genetic etiology is uncertain after standard workup. Studies onWGS report changes in management that would improve health outcomes. The effect of WGS results on health outcomes are the same as those with WES, including avoidance of invasive procedures, medication changes to reduce morbidity, discontinuation of or additional testing and initiation of palliative care or reproductive planning.

Section Summary: Whole Genome Sequencing for Children with Multiple Congenital Anomalies or a Neurodevelopmental Disorder of Unknown Etiology Following Standard Workup; Patients who are not Critically Ill

WGS has been studied in non-critically ill children with congenital abnormalities and development delays of unknown etiology following standard workup. The diagnostic yield for WGS has been reported between 20% and 40%. Additional indirect evidence is available from studies reporting diagnostic yield and change in management results of WES in a similar population, and WGS may result in similar or better diagnostic yield for pathogenic or likely pathogenic variants compared with WES although few direct comparisons are available.

Whole Genome Sequencing for a Suspected Genetic Disorder Other Than Multiple Congenital Anomalies or a Neurodevelopmental Disorder; Patients who are not Critically Ill

The purpose of WGS in patients with a suspected genetic disorder of unknown etiology following standard workup is to establish a molecular diagnosis from either the coding or noncoding regions of the genome. The criteria under which diagnostic testing for a genetic or heritable disorder may be considered clinically useful are stated above.

The question addressed in this policy is: Does WGS improve health outcomes when used for the diagnosis of patients with a suspected genetic disorder of unknown etiology following standard workup without WES or WGS?

The following PICO was used to select literature to inform this review.

Patients

The relevant population of interest is:

    • Children with a suspected genetic disorder other than multiple unexplained congenital anomalies or a neurodevelopmental disorder of unknown etiology following standard workup.
Interventions

The relevant interventions being considered include:

    • WGS with trio testing when possible
Several laboratories offer WGS as a clinical service. Medical centers may also offer WGS as a clinical service.

The median time for standard WGS is several weeks.

Note that this policy does not address the use of WGS for preimplantation genetic diagnosis or screening, prenatal (fetal) testing, or for testing of cancer cells.

Comparators

The following practice is currently being used to diagnose a suspected genetic disorder: standard clinical workup without WES or WGS.

Standard clinical workup was described in a preceding section.

Outcomes

Outcomes of interest are as described above for use of WES in patients with multiple congenital anomalies or a neurodevelopmental disorder.

Study Selection Criteria

For the evaluation of clinical validity of WGS, studies that met the following eligibility criteria were considered:

    • Reported on the diagnostic yield or performance characteristics such as sensitivity and specificity of rapid WGS or WGS;
    • Patient/sample clinical characteristics were described;
    • Patient/sample selection criteria were described;
    • Included at least 20 patients.
Technically Reliable

Assessment of technical reliability focuses on specific tests and operators and requires review of unpublished and often proprietary information. Review of specific tests, operators, and unpublished data are outside the scope of this policy and alternative sources exist. This policy focuses on the clinical validity and clinical utility.

Clinically Valid

A test must detect the presence or absence of a condition, the risk of developing a condition in the future, or treatment response (beneficial or adverse).

The use of WGS has been studied in children with a suspected genetic disorder other than multiple unexplained congenital anomalies or a neurodevelopmental disorder in several observational studies, both prospective and retrospective. Studies are described in Table 8. The diagnostic yield of WGS has been between 9% and 55%. However, these studies include mixed indications with heterogeneous populations and include little information about associated changes in management following genetic diagnosis.

Table 8. Diagnostic Yields with Whole Genome Sequencing in Children with a Suspected Genetic Disorder other than Multiple Unexplained Congenital Anomalies or a Neurodevelopmental Disorder of Unexplained Etiology Following Standard Workup
Study
Patient Population
N
Design
Yield,n (%)
Additional Information
Thiffault et al (2019)40,Patients with suspected genetic disorders referred for genetic testing between 2015 and 2017. The majority had previous genetic testing without diagnosis.The mean age was 7 yrs.80Prospective. The majority underwent trio sequencing; WGS was performed for the proband and WES was done for both parents19 (24)2 partial gene deletions detected with WGS that would not be detectable with WES
Alfares et al (2018)41,Undiagnosed patients (91% pediatric) who had a history of negative WES testing
70% Consanguinity
154 recruited; 108 included in analysisRetrospective, selection method and criteria unclear10 (9%)Reported incremental yield of WGS in patients with negative CGH and WES
Carss et al (2017)42,Unexplained inherited retinal disease; ages not specified605Retrospective NIHR-BioResource Rare Diseases Consortium331 (55)Compared with a detection rate of 50% with WES (n=117)
Ellingford et al (2016)43,Unexplained inherited retinal disease; ages not specified46Prospective WGS in patients referred to a single-center24 (52)Estimated 29% increase in yield vs. targeted NGS
Taylor et al (2015)44,Broad spectrum of suspected genetic disorders (Mendelian and immunological disorders)217Prospective, multicenter series
Clinicians and researchers submitted potential candidates for WGS and selections were made by a scientific Steering Committee. Patients were eligible if known candidate genes and large chromosomal copy number changes had been excluded.
Trio testing for a subset of 15 families.
46 (21)34% yield in Mendelian disorders; 57% yield in trios
Yuen et al (2015)45,Patients with diagnosed autism spectrum disorder50Prospective; unclear how patients were selected; quartet testing of extensively phenotyped families (parents and 2 ASD-affected siblings)21 (42%)12/20 had change in management; 1/20 had change in reproductive counseling
ASD: autism spectrum disorder;CGH: comparative genomic hybridization; NGS: next-generation sequencing; NIHR: National Institute for Health Research;WGS: whole genome sequencing; WES: whole exome sequencing;
1
SNV/indel

Tables 9 and 10 display notable limitations identified in each study.

Table 9. Relevance Limitations for Studies of Whole Genome Sequencing
Study
Populationa
Interventionb
Comparatorc
Outcomesd
Duration of Follow-Upe
Thiffault et al (2019)40,3. Included heterogeneous diseases
Alfares et al (2018)41,3: Clinical characteristics not described4: 70% consanguinity3. Appears to be proband testing only but not clear
Carss et al (2017)42,4. 25% had no prescreening performed
Ellingford et al (2016)43,3. Proband testing only
Taylor et al (2015)44,3. Included highly heterogeneous diseases
Yuen et al (2015)45,4: All patients had a clinical diagnosis3: Results of standard diagnostic methods not discussed
The study limitations stated in this table are those notable in the current review; this is not a comprehensive limitations assessment.
WGS: whole genome sequencing.

Population key: 1. Intended use population unclear; 2. Clinical context is unclear; 3. Study population is unclear; 4. Study population not representative of intended use.
b
Intervention key: 1. Classification thresholds not defined; 2. Version used unclear; 3. Not intervention of interest.
c
 Comparator key: 1. Classification thresholds not defined; 2. Not compared to credible reference standard; 3. Not compared to other tests in use for same purpose.
d
 Outcomes key: 1. Study does not directly assess a key health outcome; 2. Evidence chain or decision model not explicated; 3. Key clinical validity outcomes not reported (sensitivity, specificity, and predictive values); 4. Reclassification of diagnostic or risk categories not reported; 5. Adverse events of the test not described (excluding minor discomforts and inconvenience of venipuncture or noninvasive tests).
e
 Follow-Up key: 1. Follow-up duration not sufficient with respect to natural history of disease (true-positives, true-negatives, false-positives, false-negatives cannot be determined).

Table 10. Study Design and Conduct Limitations for Studies of Whole Genome Sequencing
Study
Selectiona
Blindingb
Delivery of Testc
Selective Reportingd
Data Completenesse
Statisticalf
Thiffault et al (2019)40,1,2: Unclear how patients were selected from those eligible
Alfares et al (2018)41,1,2: Unclear how patients were selected from those eligible
Carss et al (2017)42,
Ellingford et al (2016)43,
Taylor et al (2015)44,
Yuen et al (2015)45,1,2. Unclear how patients were selected from those eligible
The study limitations stated in this table are those notable in the current review; this is not a comprehensive limitations assessment. WGS: whole genome sequencing.
Selection key: 1. Selection not described; 2. Selection not random or consecutive (ie, convenience).
Blinding key: 1. Not blinded to results of reference or other comparator tests.
Test Delivery key: 1. Timing of delivery of index or reference test not described; 2. Timing of index and comparator tests not same; 3. Procedure for interpreting tests not described; 4. Expertise of evaluators not described.
d
 Selective Reporting key: 1. Not registered; 2. Evidence of selective reporting; 3. Evidence of selective publication.
e
 Data Completeness key: 1. Inadequate description of indeterminate and missing samples; 2. High number of samples excluded; 3. High loss to follow-up or missing data.
f
 Statistical key: 1. Confidence intervals and/or p values not reported; 2. Comparison with other tests not reported.

Clinically Useful

A test is clinically useful if the use of the results informs management decisions that improve the net health outcome of care. The net health outcome can be improved if patients receive correct therapy, or more effective therapy, or avoid unnecessary therapy, or avoid unnecessary testing.

Direct Evidence

Direct evidence of clinical utility is provided by studies that have compared health outcomes for patients managed with and without the test. Because these are intervention studies, the preferred evidence would be from RCTs.

No RCTs assessing the use of WGS to diagnose a suspected genetic disorder other than multiple unexplained congenital anomalies or a neurodevelopmental disorder were identified.

Chain of Evidence

Indirect evidence on clinical utility rests on clinical validity. If the evidence is insufficient to demonstrate test performance, no inferences can be made about clinical utility.

A genetic diagnosis for an unexplained disorder can alter management in several ways: such a diagnosis may lead to including genetic counseling and ending the diagnostic odyssey and may affect reproductive decision making.

Because the clinical validity of WGS for this indication has not been established, a chain of evidence cannot be constructed.

Section Summary: Whole Genome Sequencing for a Suspected Genetic Disorder Other Than Multiple Congenital Anomalies or a Neurodevelopmental Disorder; Patients who are not Critically Ill

WGS has also been studied in children with a suspected genetic disorder other than multiple unexplained congenital anomalies or a neurodevelopmental disorder of unknown etiology following standard workup. The diagnostic yield of WGS has been between 9% and 55%. However, these studies include mixed indications with heterogeneous populations and include little information about associated changes in management following genetic diagnosis.

Rapid Whole Exome or Genome Sequencing in Critically Ill Infants or Children

The purpose of rapid whole exome sequencing (rWES) or rapid whole genome sequencing (rWGS) in critically ill patients with a suspected genetic disorder of unknown etiology is to establish a molecular diagnosis from either the coding or noncoding regions of the genome. The criteria under which diagnostic testing for a genetic or heritable disorder may be considered clinically useful are stated above.

The most common cause of death in neonates in the United States is genetic disorders. Currently, critically ill neonates with suspected genetic diseases are frequently discharged or deceased without a diagnosis. There are thousands of rare genetic disorders. The presentation of many of these disorders in neonates may be nonspecific or differ from the presentation in older patients and the disorder may produce secondary involvement of other systems due to the fragility of the neonate that obscures the primary pathology..

The neonatal intensive care unit (NICU) treatment of suspected genetic diseases is often empirical. Rapid diagnosis is critical for delivery of interventions that reduce morbidity and mortality in genetic diseases for which treatments exist. For many genetic diseases there is no effective treatment and timely diagnosis limits futile intensive care.

The question addressed in this policy is: Does rWES or rWGS improve health outcomes when used for the diagnosis ofcritically ill infants or children with a suspected genetic disorder of unknown etiology without WES or WGS?

The following PICO was used to select literature to inform this review.

Patients

The relevant population of interest is:

    • Critically ill infants presenting with any of a variety of disorders and anomalies suspected to have a genetic basis but not explained by standard workup. For example, patients may have a phenotype that does not correspond with a specific disorder for which a genetic test targeting a specific gene is available. Specifically for critically ill infants, the population would also include patients for whom specific diagnostic tests available for that phenotype are not accessible within a reasonable timeframe. Petrikin (2018) identified the critically ill infants that are appropriate for rapid testing as meeting the following inclusion criteria: multiple congenital anomalies; abnormal laboratory test suggests a genetic disease or complex metabolic phenotype; abnormal response to standard therapy for a major underlying condition; significant hypotonia; or persistent seizures. Exclusion criteria included: an infection with normal response to therapy; isolated prematurity; isolated unconjugated hyperbilirubinemia; Hypoxic Ischemic Encephalopathy; confirmed genetic diagnosis explains illness; Isolated Transient Neonatal Tachypnea; or nonviable neonates.
Interventions

The relevant interventions being considered include:

    • rapid WES with trio testing when possible
    • rapid WGS with trio testing when possible
Several laboratories offer WES or WGS as a clinical service. Medical centers may also offer rWES or rWGS or standard WES or WGS as a clinical service.

The median time for standard WGS is several weeks. The median time-to-result for rWES or rWGS is approximately 5 days or less.

Note that this policy does not address the use of WES or WGS for preimplantation genetic diagnosis or screening, prenatal (fetal) testing, or for testing of cancer cells.

Comparators

The following practice is currently being used to diagnose a suspected genetic disorder: standard clinical workup without WES or WGS.

Standard clinical workup was described in a preceding section.

Outcomes

Outcomes of interest are as described above for use of WES in patients with multiple congenital anomalies or a neurodevelopmental disorder. For critically ill infants, rapid diagnosis is important therefore, in addition to the outcomes described in the previous section, time to diagnosis and time to discharge are also outcomes of interest.

Of course, mortality is a compelling outcome. However, many of the conditions are untreatable and diagnosis of an untreatable condition may lead to earlier transition to palliative care but may not prolong survival

Study Selection Criteria

For the evaluation of clinical validity of rWES or rWGS, studies that met the following eligibility criteria were considered:

    • Reported on the diagnostic yield or performance characteristics such as sensitivity and specificity ofrWES or rWGS;
    • Patient/sample clinical characteristics were described;
    • Patient/sample selection criteria were described;
    • Included at least 20 patients.
Technically Reliable

Assessment of technical reliability focuses on specific tests and operators and requires review of unpublished and often proprietary information. Review of specific tests, operators, and unpublished data are outside the scope of this policy and alternative sources exist. This policy focuses on the clinical validity and clinical utility.

Clinically Valid

A test must detect the presence or absence of a condition, the risk of developing a condition in the future, or treatment response (beneficial or adverse).

The use ofrWES and rWGS has been studied in critically ill children in several observational studies, both prospective and retrospective, and 1 RCT. Studies are described in Table 11. The RCT is discussed in more detail in the following ‘Clinically useful’ section. One study included only infants with cardiac defects and had a diagnostic yield of 6% with WGS. The remaining studies included phenotypically diverse but critically ill infants and had yields of between 30% and 60%.

Table 11. Diagnostic Yields With Rapid Whole Exome or Whole Genome Sequencing in Critically Ill Infants or Children With a Suspected Genetic Disorder of Unknown Etiology
Study
Patient Population
N
Design
Yield,n (%)
Additional Information
rapid WES
Wu et al (2019)46,Pediatric patients (< 18 yr old) who were critically ill (PICU; 68%) and suspected of having a genetic disease or newborns who were suspected of having a serious genetic disease after newborn screening. The primary phenotypes were neurologic (35%), cardiac (22.5%), metabolic (15%), and immunological (15%). Ages ranged from 0.2 mos to 13 yrs.40Eligibility and selection from eligible patients were unclear. Trio testing was performed.21 (52.5%)
    • Clinical management was changed for 81%: medications were recommended for 10 patients (48%), transplantation
      was advised for 5 (13%), and hospice care was suggested for 2 (5%)
Elliott et al (2019) 47,RAPIDOMICSNICU neonates with unexplained seizures, metabolic disturbances (4%), neurological disorders (28%), multiple congenital anomalies (56%), or significant physiological disturbance for which diagnosis would likely change clinical management25Patient were evaluated for enrolment by a clinical geneticist and a neonatologist and approved by the research team. Trio analysis was performed.. All patients with suspected definitely, possibly, or partially causal variants generated by rWES underwent Sanger validation15 (60%)
    • 3 additional patients diagnosed with multi-gene panel testing or chromosomal microarray analysis
    • 34 discrete and immediate medical decisions were identified for 15 of the 18 diagnosed patients
Gubbels et al (2019) 48,Infants age <6 mos admitted to ICU admission with recent presentation of seizures (20%), hypotonia (40%), multiple congenital anomalies (72%), complex metabolic phenotype (32%) or other.50New ICU admissions were triaged daily by a patient selection algorithm developed by a multidisciplinary medical team (neonatology, genetics, and neurology). whole-blood samples were collected from probands and parents for trio-based exome sequencing.29 (58%)
    • Results informed medical management changes in 24 of 29 patients. For 21 patients there was an acute impact on care: switch to comfort care, specialist referral, decision not to pursue further diagnostic testing
Stark et al (2018)49,Acutely unwell pediatric patients with suspected monogenic disorders; 22% congenital abnormalities and dysmorphic features; 43% neurometabolic disorder; 35% other.40Recruited during clinical care by the clinical genetics services at the 2 tertiary pediatric hospitals; panel of study investigators reviewed eligibility; Used singleton rWES.21 (53)
    • Clinical management changed in 12 of the 21 diagnosed patients (57%)
    • Median time to report of 16 days (range, 9 to 109)
Meng et al (2017)50,Critically ill infants within the first 100 days of life who were admitted to a tertiary care center between 2011 and 2017 and who were suspected to have genetic disorders. 208 infants were in NICU or PICU at time of sample.278 overall; 208 in NICU or PICU; 63 received rWESReferred to tertiary care; proband WES in 63%, trio WES in 14; critical trio rWES in 23%.102 (37) overall;32 (51) for rWES
    • Molecular diagnoses directly affected medical management in 53 of 102 patients (52%) overall and in 23 of 32, 72% who received rWES
rapid WGS
French et al (2019)51,Infants and children in NICU and PICU admitted between 2016 and 2018 with a possible single gene disorder. Exclusion criteria for infants included: admitted for short stay post-delivery surveillance, prematurity without additional features, babies with a clear antenatal or delivery history suggestive of a non-genetic cause and those babies where a genetic diagnosis was already made.Median age, NICU: 12 days, PICU: 24 mosOverall: 195NICU: 106PICU: 61Pediatric neurology or clinical genetics department: 28Trio WGS testing (when available) for prospective cohort of families recruited in NICU and PICU at a single site in the U.K.Overall: 40 (21)NICU: 13PICU: 25Diagnosis affected clinical management in more than 65% of cases (83% in neonates) including modification of treatments (13%) and care pathways (35% in PICU, 48% in NICU) and/or informing palliative care decisions. For at least 7cases, distinguishing between inherited and de novo variants informed reproductive decisions.VUS in 2 (1%)
Sanford et al (2019)52,Children 4 mos to 18 yrs admitted to single-center PICU between 2016 and 2018 with suspicion for an underlying monogenic disease.Median age: 3 yrsPrimary reasons for admission: respiratory failure (18%), shock (16%), altered mental status (13%), and cardiac arrest (13%)38Trio rWGS testing (when available) in retrospective cohort study of, consecutive children who had rWGS after admission to a single-center tertiary hospital in the U.S.17 (45)VUS identified in all cases but were not reported to patients.Changes in ICU management in 4diagnosed children (24%), 3 patients had medication changes, 14 children had a subacute (non-ICU) change in the clinical management had implications for family screening
Hauser et al (2018)53,Neonatal and pediatric patients born with a cardiac defect in whom the suspected genetic disorder had not been found using conventional genetic methods34TriorWGS testing for patients recruited from the NICU, PICU, or general inpatient pediatric ward of a single-center2 (6)VUS in 10 (26%)
Farnaes et al (2018)54,Critically ill infants with undiagnosed, highly diverse phenotypes. Median age 62 days (range 1-301 days).
Multiple congenital anomalies, 29%; Neurological, 21%; Hepatic, 19%
42Retrospective; comparative (receivedrWGS and standard testing (mostly commonly CMA)Trio testing (when available) usingrWGS18 (43)10% were diagnosed by standard test
Change in management after WGS in 13 of 18 (72%) patients with new genetic diagnosis
Estimated that rWGS reduced length of stay by 124 days
Mestek-Boukhibar et al (2018)55,Acutely ill infants with suspected underlying monogenetic disease. Median age 2.5 mos.
Referred from Clinical genetics, 42%; Immunology 21%; intensive care, 13%
24Prospective;rWGS trio testing in a tertiary children's hospital PICU and pediatric cardiac intensive care unit.10 (42)Change in management:In 3 patients
Van Diemen (2018)56,Critically ill infants with undiagnosed illness excluding those with clear clinical diagnosis for which a single targeted test or gene panel was available; median age 28 days.
Presentation: cardiomyopathy, 17%, severe seizure disorder, 22%, abnormal muscle tone, 26%, 13% liver failure
23ProspectiverWGS Trio testing of patients from NICU/PICU; decision to include a patient was made by a multidisciplinary team; regular genetic and other investigations were performed in parallel7 (30)2 patients required additional sequencing data
1 incidental findingWGS led to the withdrawal of unsuccessful intensive care treatment in 5 of the 7 children diagnosed
Willig (2015)57,Acutely ill infants with undiagnosed illness, suspected genetic etiology; 26% congenital anomalies; 20% neurological; 14% cardiac; 11% metabolic; Median age 26 days35Retrospective; enrolled in a research biorepository (nominated by treated physician, reviewed by panel of experts); hadrWGS and standard diagnostic tests to diagnose monogenic disorders of unknown cause; trio testing20 (57)had diagnoses with ‘strongly favorable effects on management’Palliative care initiated in 6 infantsof 20 WGS diagnoses were diseases that were not part of the differential at time of enrollment

CMA: chromosomal microarray analysis; ICU: intensive care unit; NICU: neonatal intensive care unit; PICU: pediatric intensive care unit; RAPIDOMICS: rapid genome-wide sequencing in a neonatal intensive care unit-successes and challenges; rWGS: rapid whole genome sequencing;
rWES: rapid whole exome sequencing;
WGS: whole genome sequencing; WES: whole exome sequencing; VUS: variant of uncertain significance.

Tables 12 and 13 display notable limitations identified in each study.

Table 12. Relevance Limitations for Studies of Rapid Whole Exome or Whole Genome Sequencing
Study
Populationa
Interventionb
Comparatorc
Outcomesd
Duration of Follow-Upe
Wu et al (2019)46,3: Results of standard diagnostic methods not discussed
Elliott et al (2019) 47,
Gubbels et al (2019) 48,3: Results of standard diagnostic methods not discussed
Stark et al (2018)49,3. Included highly heterogeneous diseases3. Proband testing only3: Results of standard diagnostic methods not discussed
Meng et al (2017)50,3: Not all patients received rapid testing3: Chromosomal microarray analysis was completed for 85% but results not discussed
French et al (2019)51,3: No comparator
Sanford et al (2019)52,3: No comparator
Hauser et al (2018)53,3: No comparator
Farnaes et al (2018)54,3. Included highly heterogeneous diseases
Mestek-Boukhibar et al (2018)55,3. Included highly heterogeneous diseases3: No comparator
Van Diemen (2018)56,3. Included highly heterogeneous diseases3: Results of standard diagnostic methods not discussed; were available afterrWGS
Willig et al (2015)57,3. Included highly heterogeneous diseases3: Results of standard diagnostic methods not discussed
Gilissen et al (2014)39,
The study limitationsstated in this table are those notable in the current review; this is not a comprehensive limitations assessment.
rWES: rapid whole exome sequencing;rWGS: rapid whole genome sequencing.

Population key: 1. Intended use population unclear; 2. Clinical context is unclear; 3. Study population is unclear; 4. Study population not representative of intended use.
Intervention key: 1. Classification thresholds not defined; 2. Version used unclear; 3. Not intervention of interest.
c
 Comparator key: 1. Classification thresholds not defined; 2. Not compared to credible reference standard; 3. Not compared to other tests in use for same purpose.
d
 Outcomes key: 1. Study does not directly assess a key health outcome; 2. Evidence chain or decision model not explicated; 3. Key clinical validity outcomes not reported (sensitivity, specificity and predictive values); 4. Reclassification of diagnostic or risk categories not reported; 5. Adverse events of the test not described (excluding minor discomforts and inconvenience of venipuncture or noninvasive tests).
e
 Follow-Up key: 1. Follow-up duration not sufficient with respect to natural history of disease (true-positives, true-negatives, false-positives, false-negatives cannot be determined).

Table 13. Study Design and Conduct Limitations for Studies of Rapid Whole Exome or Whole Genome Sequencing
Study
Selectiona
Blindingb
Delivery of Testc
Selective Reportingd
Data Completenesse
Statisticalf
Wu et al (2019)46,1: Criteria for selection unclear
Elliott et al (2019)47,2: Potential enrollees selected by a panel
Gubbels et al (2019)48,2: New ICU admissions were triaged 1 team and enrollment criteria were applied by a panel
Stark et al (2018)49,2: Eligibility determined by panel; a minimum of 2 clinical geneticists had to agree rWES was appropriate for a patient to be enrolled
Meng et al (2017)50,1,2 Unclear if the patients were randomly or consecutively chosen from those who were eligible
French et al (2019)51,1,2. Unclear how patients were selected from those eligible
Sanford et al (2019)52,
Hauser et al (2018)53,
Farnaes et al (2018)54,2: Patients nominated by clinicians
Mestek-Boukhibar et al (2018)55,2: Eligibility criteria established after first 10 enrolled.
Van Diemen (2018)56,2: Decision to include a patient was made by a multidisciplinary team
Willig et al (2015)57,2: Nominated by treated physician, reviewed by panel of experts for inclusion
Gilissen et al (2014)39,
The study limitationsstated in this table are those notable in the current review; this is not a comprehensive limitations assessment. rWES: rapid whole exome sequencing; rWGS: rapid whole genome sequencing.
Selection key: 1. Selection not described; 2. Selection not random or consecutive (ie, convenience).
b
Blinding key: 1. Not blinded to results of reference or other comparator tests.
c
Test Delivery key: 1. Timing of delivery of index or reference test not described; 2. Timing of index and comparator tests not same; 3. Procedure for interpreting tests not described; 4. Expertise of evaluators not described.
d
 Selective Reporting key: 1. Not registered; 2. Evidence of selective reporting; 3. Evidence of selective publication.
e
 Data Completeness key: 1. Inadequate description of indeterminate and missing samples; 2. High number of samples excluded; 3. High loss to follow-up or missing data.
f
 Statistical key: 1. Confidence intervals and/or p values not reported; 2. Comparison with other tests not reported.

Clinically Useful

A test is clinically useful if the use of the results informs management decisions that improve the net health outcome of care. The net health outcome can be improved if patients receive correct therapy, or more effective therapy, or avoid unnecessary therapy, or avoid unnecessary testing.

Direct Evidence

Direct evidence of clinical utility is provided by studies that have compared health outcomes for patients managed with and without the test. Because these are intervention studies, the preferred evidence would be from RCTs.

Kingsmore et al (2019) reported early results of A Randomized, Blinded, Prospective Study of the Clinical Utility of Rapid Genomic Sequencing for Infants in the Acute-care Setting (NSIGHT2) trial58,. NSIGHT2 was a randomized, controlled, blinded trial of the effectiveness of rapid whole-genome or -exome sequencing (rWGS or rWES, respectively) in seriously ill infants with diseases of unknown etiology primarily from the NICU, pediatric intensive care unit (PICU), and cardiovascular intensive care unit (CVICU) at a single hospital in San Diego. Details of the study are provided in Table 14and results are shown in Table 15. 95 infants were randomized to rWES and 94 to rWGS; in addition 24 infants who were gravely ill received ultrarapid whole-genome sequencing (urWGS). The initial Kingsmore et al (2019) publication included only the diagnostic outcomes. Other outcomes are expected in future publications. The registration for the study (NSIGHT2; NCT03211039) indicates that 1000 infants are expected to be enrolled; the Kingsmore et al (2019) publication does not specify whether enrollment is continuing. The diagnostic yield of rWGS and rWES was similar (19% vs. 20%, respectively), as was time (days) to result (median, 11 vs. 11 days). Although the urWGS was not part of the randomized portion of the study, the proportion diagnosed by urWGS was (11 of 24 [46%]) and time to result was a median of 4.6 days. The incremental diagnostic yield of reflexing to trio testing after inconclusive proband analysis was 0.7% (1 of 147).

Petrikin et al (2018) reported on the Prospective Randomized Trial of the Clinical Utility of Rapid Next Generation Sequencing in Acutely Ill Neonates(NSIGHT1; NCT02225522) RCT of rWGS to diagnose suspected genetic disorders in critically ill infants.59, In brief,NSIGHT1 was an investigator-initiated (funded by National Human Genome Research Institute and Eunice Kennedy Shriver National Institute of Child Health and Human Development), blinded, and pragmatic trial comparing trio rWGS with standard genetic tests to standard genetic tests alone with a primary outcome of proportion of NICU/PICU infants receiving a genetic diagnosis within 28 days. Parents of patients and clinicians were unblinded after 10 days and compassionate cross-over to rWGS occurred in 5 control patients. The study was designed to enroll 500 patients in each group but was terminated early due to loss of equipoise on the part of study clinicians who began to regard standard tests alone as inferior to standard tests plus trio rWGS. Intention-to-treat analyses were reported, ie, crossovers were included in the group to which they were randomized. The trial required confirmatory testing of WGS results which lengthened the time to rWGS diagnosis by 7–10 days. Study characteristics are shown in Table 14 and results are shown in Table 15.

Table 14. Characteristics of RCTs of Rapid Whole Genome Sequencing in Critically Ill Infants
Study; Trial
Countries
Sites
Dates
Participants
Interventions1
Active
Comparator
Kingsmore et al (2019)58,NSIGHT2 (NCT03211039)U.S.12017 to 2018Acutely ill infants, primarily from the NICU, PICU, and CVICU; age <4 mos; time from admission or time from development of a feature suggestive of a genetic condition of <96 h; excluding infants in whom there was a very low likelihood that a genetic disease diagnosis would change management.N=94, rWGS initially performed with proband sequences alone; if diagnosis was not made analysis was performed again, with parental samplesN=95, rWES initially performed with proband sequences alone; if diagnosis was not made analysis was performed again, with parental samples
Petrikin (2018)59,;NSIGHT1 (NCT02225522)U.S.12014 to 2016Infants (<4m) in the NICU/PICU with illnesses of unknown etiology and: 1. genetic test order or genetic consult; 2. major structural congenital anomaly or at least 3 minor anomalies; 3. abnormal laboratory test suggesting genetic disease; or 4. abnormal response to standard therapy for a major underlying condition.Primary system involved:CA/musculoskeletal, 35%Neurological, 25%Cardiovascular,17%Respiratory, 6%N=32
rWGS on specimens from both biological parents and affected infants simultaneously
N=33
Standard clinical testing for genetic disease etiologies was performed in infants based on physician clinical judgment, assisted by subspecialist recommendations
CA: congenital anomalies; CVICU: cardiovascular intensive care unit; NICU: neonatal intensive care unit ; NSIGHT1: Prospective Randomized Trial of the Clinical Utility of Rapid Next Generation Sequencing in Acutely Ill Neonates; NSIGHT2; A Randomized, Blinded, Prospective Study of the Clinical Utility of Rapid Genomic Sequencing for Infants in the Acute-care Setting; PICU: pediatric intensive care unit; RCT: randomized controlled trial; rWES: rapid whole exome sequencing; rWGS: rapid whole genome sequencing.

Table 15 Results of RCTs of Rapid Whole Genome Sequencing in Critically Ill Infants
Study
Diagnostic yield
Time to diagnosis
Age at at discharge
Changes in management
Mortality
Kingsmore et al (2019) 58,NSIGHT2Genetic diagnosis, timing unspecified (%)Proportion of results reported within 7 days (%)Mortality at 28 days (%)
N189189NRNR189
rWGS20%11%3%
rWES19%4%0%
Treatment effect (95% CI)p=0.88p=0.10p=0.25
Petrikin et al (2018)59,;NSIGHT1Genetic diagnosis within 28 days of enrollment (%)Time (days) to diagnosis from enrollment, medianAge (days) at hospital discharge, meanChange in management related to test results (%)Mortality at 180 days (%)
N6565656565
rWGS31%1366.341%113%
Standard testing3%10768.524%112%
Treatment effect (95% CI)p=0.003p=0.002p=0.91p=0.11NR
CI: confidence interval: RCT: randomized controlled trial; NR: not reported; NSIGHT1: Prospective Randomized Trial of the Clinical Utility of Rapid Next Generation Sequencing in Acutely Ill Neonates;
NSIGHT2; A Randomized, Blinded, Prospective Study of the Clinical Utility of Rapid Genomic Sequencing for Infants in the Acute-care Setting;
rWES: rapid whole exome sequencing; rWGS: rapid whole genome sequencing.

1
Includes changes related to positive result (diagnosis); does not include impact of negative test results on management.

Tables 16 and 17 display notable limitations identified in each study.

Table 16. Relevance Limitations of RCTs of Rapid Whole Genome Sequencing in Critically Ill Infants
Study
Populationa
Interventionb
Comparatorc
Outcomesd
Follow-Upe
Kingsmore et al (2019) 58,NSIGHT21: Initial publicaion includes only diagnostic outcomes5: No discussion of clinically significant differences1,2: Follow-up unclear
Petrikin et al (2018)59,
The study limitations stated in this table are those notable in the current review; this is not a comprehensive limitations assessment. RCT: randomized controlled trial; NSIGHT2; A Randomized, Blinded, Prospective Study of the Clinical Utility of Rapid Genomic Sequencing for Infants in the Acute-care Setting; rWGS: rapid whole genome sequencing.
Population key: 1. Intended use population unclear; 2. Clinical context is unclear; 3. Study population is unclear; 4. Study population not representative of intended use.
Intervention key: 1. Not clearly defined; 2. Version used unclear; 3. Delivery not similar intensity as comparator; 4.Not the intervention of interest.
c
 Comparator key: 1. Not clearly defined; 2. Not standard or optimal; 3. Delivery not similar intensity as intervention; 4. Not delivered effectively.
d
 Outcomes key: 1. Key health outcomes not addressed; 2. Physiologic measures, not validated surrogates; 3. No CONSORT reporting of harms; 4. Not establish and validated measurements; 5. Clinical significant difference not prespecified; 6. Clinical significant difference not supported.
e
 Follow-Up key: 1. Not sufficient duration for benefit; 2. Not sufficient duration for harms.

Table 17. Study Design and Conduct Limitations of RCTs of Rapid Whole Genome Sequencing in Critically Ill Infants
Study
Allocationa
Blindingb
Selective Reportingd
Data Completenesse
Powerd
Statisticalf
Kingsmore et al (2019)58,NSIGHT23: Allocation concealment not described1: Power calculations not reported; clinicaltrials.gov listing indicates that 1000 infants were expected but only 189 were reported in the initial report4 :Only p-values reported; no treatment effects
Petrikin et al (2018)59,1: Parents/clinicians unblinded at day 10 but analyses were intention-to-treat so crossovers would bias toward null4: Trial stopped early, power for secondary outcomes will be very low3, 4: Only p-values reported with no treatment effects or CIs
The study limitations stated in this table are those notable in the current review; this is not a comprehensive limitations assessment. CI: confidence interval; RCT: randomized controlled trial; NSIGHT2; A Randomized, Blinded, Prospective Study of the Clinical Utility of Rapid Genomic Sequencing for Infants in the Acute-care Setting; rWGS: rapid whole genome sequencing; rWGS: rapid whole genome sequencing.
Allocation key: 1. Participants not randomly allocated; 2. Allocation not concealed; 3. Allocation concealment unclear; 4. Inadequate control for selection bias.
Blinding key: 1. Not blinded to treatment assignment; 2. Not blinded outcome assessment; 3. Outcome assessed by treating physician.
c
 Selective Reporting key: 1. Not registered; 2. Evidence of selective reporting; 3. Evidence of selective publication.
d
 Data Completeness key: 1. High loss to follow-up or missing data; 2. Inadequate handling of missing data; 3. High number of crossovers; 4. Inadequate handling of crossovers; 5. Inappropriate exclusions; 6. Not intent to treat analysis (per protocol for noninferiority trials).
e
 Power key: 1. Power calculations not reported; 2. Power not calculated for primary outcome; 3. Power not based on clinically important difference; 4: Target sample size not achieved.
f
 Statistical key: 1. Analysis is not appropriate for outcome type: (a) continuous; (b) binary; (c) time to event; 2. Analysis is not appropriate for multiple observations per patient; 3. Confidence intervals and/or p values not reported; 4. Comparative treatment effects not calculated.

Chain of Evidence

Indirect evidence on clinical utility rests on clinical validity. If the evidence is insufficient to demonstrate test performance, no inferences can be made about clinical utility. Two case series with approximately 100 infants are available to estimate performance characteristics of rWES in the NICU setting.

Studies on rapid WGS report changes in management that would improve health outcomes.The effect of WGS results on health outcomes are the same as those with WES, including avoidance of invasive procedures, medication changes to reduce morbidity, discontinuation of or additional testing and initiation of palliative care or reproductive planning. A chain of evidence linking meaningful improvements in diagnostic yield and changes in management expected to improve health outcomes supports the clinical value of WGS for critically ill infants.

Section Summary: Rapid Whole Exome or Genome Sequencing in Critically Ill Infants or Children

For critically ill infants, disease may progress rapidly and genetic diagnoses must be made quickly. Results of rWES have been reported in two cases including approximately 100 infants and children. Due to the limited data available, diagnostic yield and management changes are not well characterized.

Rapid WGS has increased coverage compared to WES. One RCT comparingtriorWGS with standard genetic tests to diagnose suspected genetic disorders in critically ill infants funded by National Institutes of Health has been conducted. The study was terminated early due to loss of equipoise on the part of study clinicians who began to regard standard tests alone as inferior to standard tests plus trio rWGS. The rate of genetic diagnosis within 28 days of enrollment was higher for rWGS versus standard tests (31% vs. 3%; p=0.003) and the time to diagnosis was shorter (13 days vs. 107 days; p=0.002). The age at hospital discharge and mortality rates were similar in the 2 groups. However, many of the conditions are untreatable and diagnosis of an untreatable condition may lead to earlier transition to palliative care but may not prolong survival. A second RCT compared rWGS to rWES in seriously ill infants with diseases of unknown etiology from the NICU, PICU, and CVICU. Only the diagnostic outcomes have currently been reported. The diagnostic yield of rWGS and rWES was similar (19% vs. 20%, respectively), as was time (days) to result (median, 11 vs. 11 days).. Several retrospective and prospective observational studies with sample sizes ranging from about 20 to more than 275 and in total including more than 450 critically ill infants or children reporting on diagnostic yield for rWGS ior rWES included phenotypically diverse but critically ill infants and had yields of between 30% and 60% and reports of changes in management such as avoidance of invasive procedures, medication changes, discontinuation of or additional testing and initiation of palliative care.

Summary of Evidence

For individuals who are children who are not critically ill with multiple unexplained congenital anomalies or a neurodevelopmental disorder of unknown etiology following standard workup who receive WES with trio testing when possible, the evidence includes large case series and within-subject comparisons. Relevant outcomes are test validity, functional outcomes, changes in reproductive decision making, and resource utilization. Patients who have multiple congenital anomalies or a developmental disorder with a suspected genetic etiology, but whose specific genetic alteration is unclear or unidentified by standard clinical workup, may be left without a clinical diagnosis of their disorder, despite a lengthy diagnostic workup. For a substantial proportion of these patients, WES may return a likely pathogenic variant. Several large and smaller series have reported diagnostic yields of WES ranging from 25% to 60%, depending on the individual’s age, phenotype, and previous workup. One comparative study found a 44% increase in yield compared with standard testing strategies. Many of the studies have also reported changes in patient management, including medication changes, discontinuation of or additional testing, ending the diagnostic odyssey, and family planning. The evidence is sufficient to determine that the technology results in a meaningful improvement in the net health outcome.

For individuals who are children with a suspected genetic disorder other than multiple congenital anomalies or a neurodevelopmental disorder of unknown etiology following standard workup who receive WES with trio testing when possible, the evidence includes small case series and prospective research studies. Relevant outcomes are test validity, functional outcomes, changes in reproductive decision making, and resource utilization. There is an increasing number of reports evaluating the use of WES to identify a molecular basis for disorders other than multiple congenital anomalies or neurodevelopmental disorders. The diagnostic yields in these studies range from as low as 3% to 60%. Some studies have reported on the use of a virtual gene panel with restricted analysis of disease-associated genes, and WES data allows reanalysis as new genes are linked to the patient phenotype. Overall, a limited number of patients have been studied for any specific disorder, and clinical use of WES for these disorders is at an early stage with uncertainty about changes in patient management. The evidence is insufficient to determine the effects of the technology on health outcomes.

For individuals who are children who are not critically ill with multiple unexplained congenital anomalies or a neurodevelopmental disorder of unknown etiology following standard workup who receive WGS with trio testing when possible, the evidence includes case series. Relevant outcomes are test validity, functional outcomes, changes in reproductive decision making, and resource utilization. In studies of children with congenital abnormalities and development delays of unknown etiology following standard clinical workup, the yield of WGS has been between 20% and 40%. Additional indirect evidence is available from studies reporting diagnostic yield and change in management results of WES in a similar population. WGS may result in similar or better diagnostic yield for pathogenic or likely pathogenic variants as compared with WES but few direct comparisons are available. The evidence is insufficient to determine the effects of the technology on health outcomes..

For individuals who are children with a suspected genetic disorder other than multiple unexplained congenital anomalies or a neurodevelopmental disorder of unknown etiology following standard workup who receive who receive WGS with trio testing when possible, the evidence includes case series. Relevant outcomes are test validity, functional outcomes, changes in reproductive decision making, and resource utilization. WGS has also been studied in other genetic conditions with yield ranging from 9% to 55%. Overall, a limited number of patients have been studied for any specific disorder, and clinical use of WGS as well as information regarding meaningful changes in management for these disorders is at an early stage. The evidence is insufficient to determine the effects of the technology on health outcomes.

For individuals who are critically ill infants with a suspected genetic disorder of unknown etiology following standard workup who receive rapid WGS (rWGS) or rapid WES (rWES) with trio testing when possible, the evidence includes randomized controlled trials (RCTs) and case series. Relevant outcomes are test validity, functional outcomes, changes in reproductive decision making, and resource utilization. One RCT comparing rapid trio WGS (rWGS) with standard genetic tests to diagnose suspected genetic disorders in critically ill infants was terminated early due to loss of equipoise. The rate of genetic diagnosis within 28 days of enrollment was higher for rWGS versus standard tests (31% vs. 3%; p=0.003). Changes in management due to test results were reported in 41% vs. 21% (p=0.11) of rWGS vs control patients; however, 73% of control subjects received broad genetic tests (eg, next-generation sequencing panel testing, WES, or WGS) as part of standard testing. A second RCT compared rWGS to rWES in seriously ill infants with diseases of unknown etiology from the neonatal intensive care unit, pediatric intensive care unit, and cardiovascular intensive care unit. Only the diagnostic outcomes have currently been reported. The diagnostic yield of rWGS and rWES was similar (19% vs. 20%, respectively), as was time (days) to result (median, 11 vs. 11 days). Several retrospective and prospective studies including more than 800 critically ill infants and children in total have reported on diagnostic yield for rWGS or rWES including phenotypically diverse but critically ill infants and had yields of between 30% and 60% for pathogenic or likely pathogenic variants. Studies have also reported associated changes in patient management for patients receiving a diagnosis from rWGS or rWES, including avoidance of invasive procedures, medication changes to reduce morbidity, discontinuation of or additional testing and initiation of palliative care or reproductive planning. A chain of evidence linking meaningful improvements in diagnostic yield and changes in management expected to improve health outcomes supports the clinical value of rWGS or rWES. The evidence is sufficient to determine that the technology results in a meaningful improvement in the net health outcome.

SUPPLEMENTAL INFORMATION

Practice Guidelines and Position Statements

American College of Medical Genetics and Genomics

In 2012, the American College of Medical Genetics and Genomics (ACMG) has recommended that diagnostic testing with whole exome sequencing (WES) and whole genome sequencing (WGS) should be considered in the clinical diagnostic assessment of a phenotypically affected individual when60,:

    a. "The phenotype or family history data strongly implicate a genetic etiology, but the phenotype does not correspond with a specific disorder for which a genetic test targeting a specific gene is available on a clinical basis.
    b. A patient presents with a defined genetic disorder that demonstrates a high degree of genetic heterogeneity, making WES or WGS analysis of multiple genes simultaneously a more practical approach.
    c. A patient presents with a likely genetic disorder but specific genetic tests available for that phenotype have failed to arrive at a diagnosis.
    d. A fetus with a likely genetic disorder in which specific genetic tests, including targeted sequencing tests, available for that phenotype have failed to arrive at a diagnosis."

ACMG has recommended that for screening purposes:

WGS/WES may be considered in preconception carrier screening, using a strategy to focus on genetic variants known to be associated with significant phenotypes in homozygous or hemizygous progeny.

ACMG has also recommended that WGS and WES not be used at this time as an approach to prenatal screening or as a first-tier approach for newborn screening.

In 2014, ACMG guidelines on the clinical evaluation and etiologic diagnosis of hearing loss stated that for individuals with findings suggestive of a syndromic genetic etiology for hearing loss, “pretest genetic counseling should be provided, and, with patient’s informed consent, genetic testing, if available, should be ordered to confirm the diagnosis—this testing may include single-gene tests, hearing loss sequencing panels, WES, WGS, chromosome analysis, or microarray-based copy number analysis, depending on clinical findings.”61,

In 2016, ACMG updated its recommendations on reporting incidental findings in WGS and WES testing.62, ACMG determined that reporting some incidental findings would likely have medical benefit for the patients and families of patients undergoing clinical sequencing, recommending that, when a report is issued for clinically indicated exome and genome sequencing, a minimum list of conditions, genes, and variants should be routinely evaluated and reported to the ordering clinician. The 2016 update added 4 genes and removed 1 gene resulting in an updated secondary findings minimum list including 59 medically actionable genes recommended for return in clinical genomic sequencing.

American Academy of Neurology et al

In 2014, the American Academy of Neurology and American Association of Neuromuscular and Electrodiagnostic Medicine issued evidence-based guidelines on the diagnosis and treatment of limb-girdle and distal dystrophies, which made the following recommendations (see Table 18).63,

Table 18. Guidelines on Limb-Girdle Muscular Dystrophy
Recommendation
LOE
Diagnosis
    • For patients with suspected muscular dystrophy, clinicians should use a clinical approach to guide genetic diagnosis based on the clinical phenotype, including the pattern of muscle involvement, inheritance pattern, age at onset, and associated manifestations (eg, early contractures, cardiac or respiratory involvement).
B
    • In patients with suspected muscular dystrophy in whom initial clinically directed genetic testing does not provide a diagnosis, clinicians may obtain genetic consultation or perform parallel sequencing of targeted exomes, whole-exome sequencing, whole-genome screening, or next-generation sequencing to identify the genetic abnormality.
C
Management of cardiac complications
    • Clinicians should refer newly diagnosed patients with (1) limb-girdle muscular dystrophy (LGMD)1A, LGMD1B, LGMD1D, LGMD1E, LGMD2C–K, LGMD2M–P, … or (2) muscular dystrophy without a specific genetic diagnosis for cardiology evaluation, including electrocardiogram (ECG) and structural evaluation (echocardiography or cardiac magnetic resonance imaging [MRI]), even if they are asymptomatic from a cardiac standpoint, to guide appropriate management.
B
    • If ECG or structural cardiac evaluation (eg, echocardiography) has abnormal results, or if the patient has episodes of syncope, near-syncope, or palpitations, clinicians should order rhythm evaluation (eg, Holter monitor or event monitor) to guide appropriate management.
B
    • Clinicians should refer muscular dystrophy patients with palpitations, symptomatic or asymptomatic tachycardia or arrhythmias, or signs and symptoms of cardiac failure for cardiology evaluation.
B
    • It is not obligatory for clinicians to refer patients with LGMD2A, LGMD2B, and LGMD2L for cardiac evaluation unless they develop overt cardiac signs or symptoms.
B
Management of pulmonary complications
    • Clinicians should order pulmonary function testing (spirometry and maximal inspiratory/expiratory force in the upright and, if normal, supine positions) or refer for pulmonary evaluation (to identify and treat respiratory insufficiency) in muscular dystrophy patients at the time of diagnosis, or if they develop pulmonary symptoms later in their course.
B
    • In patients with a known high risk of respiratory failure (eg, those with LGMD2I …), clinicians should obtain periodic pulmonary function testing (spirometry and maximal inspiratory/expiratory force in the upright position and, if normal, in the supine position) or evaluation by a pulmonologist to identify and treat respiratory insufficiency.
B
    • It is not obligatory for clinicians to refer patients with LGMD2B and LGMD2L for pulmonary evaluation unless they are symptomatic.
C
    • Clinicians should refer muscular dystrophy patients with excessive daytime somnolence, nonrestorative sleep (eg, frequent nocturnal arousals, morning headaches, excessive daytime fatigue), or respiratory insufficiency based on pulmonary function tests for pulmonary or sleep medicine consultation for consideration of noninvasive ventilation to improve quality of life.
B
LOE: level of evidence; LGMD: limb-girdle muscular dystrophy.

U.S. Preventive Services Task Force Recommendations

Not applicable.

Ongoing and Unpublished Clinical Trials

Some currently unpublished trials that might influence this review are listed in Table 19.

Table 19. Summary of Key Trials
NCT No.
Trial Name
Planned Enrollment
Completion Date
Ongoing
NCT02826694North Carolina Newborn Exome Sequencing for Universal Screening
400
Jun 2019(ongoing)
NCT03211039Prenatal Precision Medicine (NSIGHT2): A Randomized, Blinded, Prospective Study of the Clinical Utility of Rapid Genomic Sequencing for Infants in the Acute-care Setting
1000
Aug 2019
NCT03290469NICUSeq: A Prospective Trial to Evaluate the Clinical Utility of Human Whole Genome Sequencing (WGS) Compared to Standard of Care in Acute Care Neonates and Infants
355
Jul 2019
NCT02699190LeukoSEQ: Whole Genome Sequencing as a First-Line Diagnostic Tool for Leukodystrophies
450
Aug 2020
NCT03829176Investigating the Feasibility and Implementation of Whole Genome Sequencing in Patients With Suspected Genetic Disorder
200
Jun 2020
NCT02422511Genomic Sequencing for Childhood Risk and Newborn Illness (The BabySeq Project)
1440
Apr 2020
NCT03525431Genomic Sequencing to Aid Diagnosis in Pediatric and Prenatal Practice: Examining Clinical Utility, Ethical Implications, Payer Coverage, and Data Integration in a Diverse Population
800
May 2021
NCT03548779North Carolina Genomic Evaluation by Next-generation Exome Sequencing, 2
1700
May 2021
NCT03918707Utility of Rapid Whole Genome Sequencing in the NICU: A Pilot Study
115
Jan 2022
NCT01736566The MedSeq Project Pilot Study: Integrating Whole Genome Sequencing Into the Practice of Clinical Medicine
213
Apr 2022
NCT04170985NeuroSeq: A Prospective Trial to Evaluate the Diagnostic Yield of Human Whole Genome Sequencing (WGS) Compared to Standard of Care in Adults With Suspected Genetic Neurological Disorders
100
Jun 2022
NCT04154891Genome Sequencing Strategies for Genetics Diagnosis of Patients With Intellectual Disability (DEFIDIAG)
3825
Nov 2023
NCT03632239The Genomic Ascertainment Cohort (TGAC)
1000
Dec 2028
NCT00410241ClinSeq: A Large-Scale Medical Sequencing Clinical Research Pilot Study
2650
Not reported
NCT03385876Rapid Whole Genome Sequencing (rWGS): Rapid Genomic Sequencing for Acutely Ill Patients and the Collection, Storage, Analysis, and Distribution of Biological Samples, Genomic and Clinical Data
100000
Dec 2050
Unpublished
NCT02380729Mutation Exploration in Non-acquired, Genetic Disorders and Its Impact on Health Economy and Life Quality
200
Dec 2017
(completed)
NCT: national clinical trial.]
________________________________________________________________________________________

Horizon BCBSNJ Medical Policy Development Process:

This Horizon BCBSNJ Medical Policy (the “Medical Policy”) has been developed by Horizon BCBSNJ’s Medical Policy Committee (the “Committee”) consistent with generally accepted standards of medical practice, and reflects Horizon BCBSNJ’s view of the subject health care services, supplies or procedures, and in what circumstances they are deemed to be medically necessary or experimental/ investigational in nature. This Medical Policy also considers whether and to what degree the subject health care services, supplies or procedures are clinically appropriate, in terms of type, frequency, extent, site and duration and if they are considered effective for the illnesses, injuries or diseases discussed. Where relevant, this Medical Policy considers whether the subject health care services, supplies or procedures are being requested primarily for the convenience of the covered person or the health care provider. It may also consider whether the services, supplies or procedures are more costly than an alternative service or sequence of services, supplies or procedures that are at least as likely to produce equivalent therapeutic or diagnostic results as to the diagnosis or treatment of the relevant illness, injury or disease. In reaching its conclusion regarding what it considers to be the generally accepted standards of medical practice, the Committee reviews and considers the following: all credible scientific evidence published in peer-reviewed medical literature generally recognized by the relevant medical community, physician and health care provider specialty society recommendations, the views of physicians and health care providers practicing in relevant clinical areas (including, but not limited to, the prevailing opinion within the appropriate specialty) and any other relevant factor as determined by applicable State and Federal laws and regulations.

___________________________________________________________________________________________________________________________

Index:
Whole Exome and Whole Genome Sequencing for Diagnosis of Genetic Disorders
Whole Exome and Whole Genome Sequencing for Diagnosis of Patients with Suspected Genetic Disorders
Whole Exome Sequencing
Exome Sequencing
Whole Genome Sequencing
Genome Sequencing
TruGenome Undiagnosed Disease Test
TruGenome Predisposition Screen
TruGenome Technical Sequence Data

References:
1. Dixon-Salazar TJ, Silhavy JL, Udpa N, et al. Exome sequencing can improve diagnosis and alter patient management. Sci Transl Med. Jun 13 2012;4(138):138ra178. PMID 22700954

2. Richards S, Aziz N, Bale S, et al. 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. May 2015;17(5):405-424. PMID 25741868

3. Blue Cross and Blue Shield Association Technology Evaluation Center (TEC). Special Report: Exome Sequencing for Clinical Diagnosis of Patients with Suspected Genetic Disorders. TEC Assessments. 2013;Volume 28:Tab 3.

4. Smith HS, Swint JM, Lalani SR, et al. Clinical Application of Genome and Exome Sequencing as a Diagnostic Tool for Pediatric Patients: a Scoping Review of the Literature. Genet. Med., 2018 May 16;21(1). PMID 29760485

5. Vissers L, van Nimwegen KJM, Schieving JH, et al. A clinical utility study of exome sequencing versus conventional genetic testing in pediatric neurology. Genet Med. Sep 2017;19(9):1055-1063. PMID 28333917

6. Cordoba M, Rodriguez-Quiroga SA, Vega PA, et al. Whole exome sequencing in neurogenetic odysseys: An effective, cost- and time-saving diagnostic approach. PLoS ONE, 2018 Feb 2;13(2). PMID 29389947

7. 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 Mar 30;20(12). PMID 29595814

8. Powis Z, Farwell Hagman KD, Speare V, et al. Exome sequencing in neonates: diagnostic rates, characteristics, and time to diagnosis. Genet. Med., 2018 Mar 23;20(11). PMID 29565416

9. Wright CF, McRae JF, Clayton S, et al. Making new genetic diagnoses with old data: iterative reanalysis and reporting from genome-wide data in 1,133 families with developmental disorders. Genet Med. Jan 11 2018. PMID 29323667

10. Wright CF, Fitzgerald TW, Jones WD, et al. Genetic diagnosis of developmental disorders in the DDD study: a scalable analysis of genome-wide research data. Lancet. Apr 4 2015;385(9975):1305-1314. PMID 25529582

11. Nambot S, Thevenon J, Kuentz P, et al. Clinical whole-exome sequencing for the diagnosis of rare disorders with congenital anomalies and/or intellectual disability: substantial interest of prospective annual reanalysis. Genet Med. Jun 2018;20(6):645-654. PMID 29095811

12. Tsuchida N, Nakashima M, Kato M, et al. Detection of copy number variations in epilepsy using exome data. Clin Genet. Mar 2018;93(3):577-587. PMID 28940419

13. Evers C, Staufner C, Granzow M, et al. Impact of clinical exomes in neurodevelopmental and neurometabolic disorders. Mol Genet Metab. Aug 2017;121(4):297-307. PMID 28688840

14. Nolan D, Carlson M. Whole exome sequencing in pediatric neurology patients: clinical implications and estimated cost analysis. J Child Neurol. Jun 2016;31(7):887-894. PMID 26863999

15. Allen NM, Conroy J, Shahwan A, et al. Unexplained early onset epileptic encephalopathy: Exome screening and phenotype expansion. Epilepsia. Jan 2016;57(1):e12-17. PMID 26648591

16. 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. Nov 2016;18(11):1090-1096. PMID 26938784

17. Tarailo-Graovac M, Shyr C, Ross CJ, et al. Exome sequencing and the management of neurometabolic disorders. N Engl J Med. Jun 9 2016;374(23):2246-2255. PMID 27276562

18. 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. Jul 2015;17(7):578-586. PMID 25356970

19. Yang Y, Muzny DM, Xia F, et al. Molecular findings among patients referred for clinical whole-exome sequencing. JAMA. Nov 12 2014;312(18):1870-1879. PMID 25326635

20. Lee H, Deignan JL, Dorrani N, et al. Clinical exome sequencing for genetic identification of rare Mendelian disorders. JAMA. Nov 12 2014;312(18):1880-1887. PMID 25326637

21. Iglesias A, Anyane-Yeboa K, Wynn J, et al. The usefulness of whole-exome sequencing in routine clinical practice. Genet Med. Dec 2014;16(12):922-931. PMID 24901346

22. Soden SE, Saunders CJ, Willig LK, et al. Effectiveness of exome and genome sequencing guided by acuity of illness for diagnosis of neurodevelopmental disorders. Sci Transl Med. Dec 3 2014;6(265):265ra168. PMID 25473036

23. Srivastava S, Cohen JS, Vernon H, et al. Clinical whole exome sequencing in child neurology practice. Ann Neurol. Oct 2014;76(4):473-483. PMID 25131622

24. Yang Y, Muzny DM, Reid JG, et al. Clinical whole-exome sequencing for the diagnosis of Mendelian disorders. N Engl J Med. Oct 17 2013;369(16):1502-1511. PMID 24088041

25. Hauer NN, Popp B, Schoeller E, et al. Clinical relevance of systematic phenotyping and exome sequencing in patients with short stature. Genet Med. Jun 2018;20(6):630-638. PMID 29758562

26. Rossi M, El-Khechen D, Black MH, et al. Outcomes of diagnostic exome sequencing in patients with diagnosed or suspected autism spectrum disorders. Pediatr Neurol. May 2017;70:34-43.e32. PMID 28330790

27. Walsh M, Bell KM, Chong B, et al. Diagnostic and cost utility of whole exome sequencing in peripheral neuropathy. Ann Clin Transl Neurol. May 2017;4(5):318-325. PMID 28491899

28. Miller KA, Twigg SR, McGowan SJ, et al. Diagnostic value of exome and whole genome sequencing in craniosynostosis. J Med Genet. Apr 2017;54(4):260-268. PMID 27884935

29. Posey JE, Rosenfeld JA, James RA, et al. Molecular diagnostic experience of whole-exome sequencing in adult patients. Genet Med. Jul 2016;18(7):678-685. PMID 26633545

30. Ghaoui R, Cooper ST, Lek M, et al. Use of whole-exome sequencing for diagnosis of limb-girdle muscular dystrophy: outcomes and lessons learned. JAMA Neurol. Dec 2015;72(12):1424-1432. PMID 26436962

31. Valencia CA, Husami A, Holle J, et al. Clinical impact and cost-effectiveness of whole exome sequencing as a diagnostic tool: a pediatric center's experience. Front Pediatr. Aug 2015;3:67. PMID 26284228

32. Wortmann SB, Koolen DA, Smeitink JA, et al. Whole exome sequencing of suspected mitochondrial patients in clinical practice.J Inherit Metab Dis. May 2015;38(3):437-443. PMID 25735936

33. Neveling K, Feenstra I, Gilissen C, et al. A post-hoc comparison of the utility of Sanger sequencing and exome sequencing for the diagnosis of heterogeneous diseases. Hum Mutat. Dec 2013;34(12):1721-1726. PMID 24123792

34. Lionel AC, Costain G, Monfared N, et al. Improved diagnostic yield compared with targeted gene sequencing panels suggests a role for whole-genome sequencing as a first-tier genetic test. Genet Med. Apr 2018;20(4):435-443. PMID 28771251

35. Costain G, Jobling R, Walker S, et al. Periodic reanalysis of whole-genome sequencing data enhances the diagnostic advantage over standard clinical genetic testing. Eur J Hum Genet. May 2018;26(5):740-744. PMID 29453418

36. Stavropoulos DJ, Merico D, Jobling R, et al. Whole Genome Sequencing Expands Diagnostic Utility and Improves Clinical Management in Pediatric Medicine. NPJ Genom Med. Jan 13 2016;1. PMID 28567303

37. Hiatt SM, Amaral MD, Bowling KM, et al. Systematic reanalysis of genomic data improves quality of variant interpretation. Clin. Genet. 2018 Jul;94(1). PMID 29652076

38. Bowling KM, Thompson ML, Amaral MD, et al. Genomic diagnosis for children with intellectual disability and/or developmental delay. Genome Med. May 30 2017;9(1):43. PMID 28554332

39. Gilissen C, Hehir-Kwa JY, Thung DT, et al. Genome sequencing identifies major causes of severe intellectual disability. Nature. Jul 17 2014;511(7509):344-347. PMID 24896178

40. Thiffault I, Farrow E, Zellmer L, et al. Clinical genome sequencing in an unbiased pediatric cohort. Genet. Med., 2018 Jul 17;21(2). PMID 30008475

41. 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. Nov 2018;20(11):1328-1333. PMID 29565419

42. Carss KJ, Arno G, Erwood M, et al. Comprehensive rare variant analysis via whole-genome sequencing to determine the molecular pathology of inherited retinal disease. Am J Hum Genet. Jan 05 2017;100(1):75-90. PMID 28041643

43. Ellingford JM, Barton S, Bhaskar S, et al. Whole genome sequencing increases molecular diagnostic yield compared with current diagnostic testing for inherited retinal disease. Ophthalmology. May 2016;123(5):1143-1150. PMID 26872967

44. Taylor JC, Martin HC, Lise S, et al. Factors influencing success of clinical genome sequencing across a broad spectrum of disorders. Nat Genet. Jul 2015;47(7):717-726. PMID 25985138

45. Yuen RK, Thiruvahindrapuram B, Merico D, et al. Whole-genome sequencing of quartet families with autism spectrum disorder. Nat Med. Feb 2015;21(2):185-191. PMID 25621899

46. Wu ET, Hwu WL, Chien YH, et al. Critical Trio Exome Benefits In-Time Decision-Making for Pediatric Patients With Severe Illnesses. Pediatr Crit Care Med. 2019 Nov;20(11). PMID 31261230

47. 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 Aug;178(8). PMID 31172278

48. Gubbels CS, VanNoy GE, Madden JA, et al. Prospective, phenotype-driven selection of critically ill neonates for rapid exome sequencing is associated with high diagnostic yield. Genet. Med. 2019 Nov. PMID 31780822

49. Stark Z, Lunke S, Brett GR, et al. Meeting the challenges of implementing rapid genomic testing in acute pediatric care. Genet Med. Mar 15 2018. PMID 29543227

50. Meng L, Pammi M, Saronwala A, et al. Use of Exome Sequencing for Infants in Intensive Care Units: Ascertainment of Severe Single-Gene Disorders and Effect on Medical Management. JAMA Pediatr. Dec 4 2017;171(12):e173438. PMID 28973083

51. French CE, Delon I, Dolling H, et al. Whole genome sequencing reveals that genetic conditions are frequent in intensively ill children. Intensive Care Med, 2019 Mar 9;45(5). PMID 30847515

52. Sanford EF, Clark MM, Farnaes L, et al. Rapid Whole Genome Sequencing Has Clinical Utility in Children in the PICU. Pediatr Crit Care Med, 2019 Jun 28. PMID 31246743

53. Hauser NS, Solomon BD, Vilboux T, et al. Experience with genomic sequencing in pediatric patients with congenital cardiac defects in a large community hospital. Mol Genet Genomic Med. Mar 2018;6(2):200-212. PMID 29368431

54. Farnaes L, Hildreth A, Sweeney NM, et al. Rapid whole-genome sequencing decreases infant morbidity and cost of hospitalization. NPJ Genom Med. 2018;3:10. PMID 29644095

55. Mestek-Boukhibar L, Clement E, Jones WD, et al. Rapid Paediatric Sequencing (RaPS): comprehensive real-life workflow for rapid diagnosis of critically ill children. J Med Genet. Nov 2018;55(11):721-728. PMID 30049826

56. van Diemen CC, Kerstjens-Frederikse WS, Bergman KA, et al. Rapid Targeted Genomics in Critically Ill Newborns. Pediatrics. Oct 2017;140(4). PMID 28939701

57. Willig LK, Petrikin JE, Smith LD, et al. Whole-genome sequencing for identification of Mendelian disorders in critically ill infants: a retrospective analysis of diagnostic and clinical findings.Lancet Respir Med. May 2015;3(5):377-387. PMID 25937001

58. Kingsmore SF, Cakici JA, Clark MM, et al. A Randomized, Controlled Trial of the Analytic and Diagnostic Performance of Singleton and Trio, Rapid Genome and Exome Sequencing in Ill Infants. Am. J. Hum. Genet. 2019 Oct;105(4). PMID 31564432

59. Petrikin JE, Cakici JA, Clark MM, et al. The NSIGHT1-randomized controlled trial: rapid whole-genome sequencing for accelerated etiologic diagnosis in critically ill infants.NPJ Genom Med. 2018;3:6. PMID 29449963

60. ACMG Board of Directors. Points to consider in the clinical application of genomic sequencing.Genet Med. Aug 2012;14(8):759-761. PMID 22863877

61. Alford RL, Arnos KS, Fox M, et al. American College of Medical Genetics and Genomics guideline for the clinical evaluation and etiologic diagnosis of hearing loss.Genet Med. Apr 2014;16(4):347-355. PMID 24651602

62. 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. Feb 2017;19(2):249-255. PMID 27854360

63. Narayanaswami P, Weiss M, Selcen D, et al. Evidence-based guideline summary: diagnosis and treatment of limb-girdle and distal dystrophies: report of the guideline development subcommittee of the American Academy of Neurology and the practice issues review panel of the American Association of Neuromuscular & Electrodiagnostic Medicine. Neurology. Oct 14 2014;83(16):1453-1463. PMID 25313375

Codes:
(The list of codes is not intended to be all-inclusive and is included below for informational purposes only. Inclusion or exclusion of a procedure, diagnosis, drug or device code(s) does not constitute or imply authorization, certification, approval, offer of coverage or guarantee of payment.)

CPT*

    81415
    81416
    81417
    81425
    81426
    81427
    0036U

HCPCS

* CPT only copyright 2020 American Medical Association. All rights reserved. CPT is a registered trademark of the American Medical Association.

_________________________________________________________________________________________

Medical policies can be highly technical and are designed for use by the Horizon BCBSNJ professional staff in making coverage determinations. Members referring to this policy should discuss it with their treating physician, and should refer to their specific benefit plan for the terms, conditions, limitations and exclusions of their coverage.

The Horizon BCBSNJ Medical Policy Manual is proprietary. It is to be used only as authorized by Horizon BCBSNJ and its affiliates. The contents of this Medical Policy are not to be copied, reproduced or circulated to other parties without the express written consent of Horizon BCBSNJ. The contents of this Medical Policy may be updated or changed without notice, unless otherwise required by law and/or regulation. However, benefit determinations are made in the context of medical policies existing at the time of the decision and are not subject to later revision as the result of a change in medical policy

____________________________________________________________________________________________________________________________