<<

Corporate Medical Policy Whole Whole AHS – M2032 “Notification” File Name: whole_genome_whole_exome_sequencing Origination: 01/01/2019 Last CAP Review: N/A Next CAP Review: 01/01/2020 Last Review: 01/01/2019 Policy Effective April 1, 2019

Description of Procedure or Service

Definitions (WGS) is the strategy of using next-generation technology to sequence entire of a genome. Whole (WES) refers to sequencing of the exome, or coding, regions of a genome (Rabbani, et. al., 2014; NIH Intramural Sequencing Center, 2015). Next generation sequencing involves sequencing of multiple small fragments of DNA in parallel, producing fast, accurate sequencing results.

***Note: This Medical Policy is complex and technical. For questions concerning the technical language and/or specific clinical indications for its use, please consult your .

Policy BCBSNC will provide for whole exome sequencing when it is determined to be medically necessary because the medical criteria and guidelines shown below are met.

Whole Genome Sequencing is considered investigational for all indications. BCBSNC does not provide coverage for investigational services or procedures.

Benefits Application This medical policy relates only to the services or supplies described herein. Please refer to the Member's Benefit Booklet for availability of benefits. Member's benefits may vary according to benefit design; therefore member benefit language should be reviewed before applying the terms of this medical policy.

When Whole Genome, Whole Exome Sequencing is covered Whole exome sequencing is considered medically necessary for the evaluation unexplained congenital or neurodevelopmental disorder in children when all the following criteria are met:

A. The patient has been evaluated by a board-certified clinician with expertise in clinical and counseled about the potential risks of B. WES results will directly impact patient management and clinical outcome for the individual being tested C. A genetic etiology is the most likely explanation for the

Page 1 of 9 An Independent Licensee of the Blue Cross and Blue Shield Association

Whole Genome Whole Exome Sequencing AHS – M2032 “Notification” D. No other causative circumstances (e.g. environmental exposures, injury, ) can explain the symptoms E. Clinical presentation does not fit a well-described for which single- or targeted panel testing (e.g., comparative genomic hybridization/chromosomal microarray analysis) is available F. The differential diagnosis list and/or phenotype warrant testing of multiple and ONE of the following: i. WES is more practical than the separate single gene tests or panels that would be recommended based on the differential diagnosis ii. WES results may preclude the need for multiple and/or invasive procedures, follow-up, or screening that would be recommended in the absence of testing

When is not covered If whole exome sequencing has been previously performed, further genetic tests involving only exome analyses is considered investigational.

Whole exome sequencing is considered investigational for all other indications, including but not limited to, tumor sequencing.

Whole genome sequencing is considered is considered investigational for all indications.

Policy Guidelines Background Whole genome and whole exome sequencing technologies provide insight into genetic organization. Technological advances allows for DNA sequencing to be completed quickly, with sequences of huge numbers of different DNA strands completed at once. Rabbani (2014) states, “Whole-exome sequencing provides coverage of more than 95% of the , which contains 85% of disease-causing in Mendelian disorders and many disease-predisposing SNPs throughout the genome. The role of more than 150 genes has been distinguished by means of WES, and this statistics is quickly growing. In this review, the impacts of WES in as well as its consequences leading to improve are summarized.”

Whole exome sequencing provides several advantages over WGS. Although the exome portion of a gene represents 1.5-2% of the genome, it contains over 85 percent of known disease-causing mutations. This results in decreased cost as well as easier interpretation of results (Hulick, et al., 2017).

Clinical Validity and Utility

Lee et al (2014) reported on the initial clinical indications for clinical exome sequencing (CES) referrals and molecular diagnostic rates for different indications and for different test types. CES was performed on a large (n=814) single-center cohort of patients with undiagnosed, suspected genetic conditions who underwent WES. Clinical exome sequencing was conducted using the trio-CES technique that involves both parents and their affected child sequenced simultaneously. Overall, a molecular diagnosis with a causative variant in a well-established clinical gene was provided for 213/814 (26%) cases. The trio-CES was associated with higher molecular diagnostic yield (31%; 127 of 410 cases) than proband-CES or traditional molecular diagnostic methods. The investigators concluded that “additional studies designed to validate these findings and to explore the effect of this approach on clinical and economic outcomes are warranted.”

Page 2 of 9 An Independent Licensee of the Blue Cross and Blue Shield Association

Whole Genome Whole Exome Sequencing AHS – M2032 “Notification” Yang et al (2014) conducted a single-center observational study of 2000 patients with clinical whole-exome sequencing performed for a suspected . A molecular diagnosis was reported for 504 patients (25.2%) with 58% of the diagnostic mutations not previously reported. The investigators concluded that “the yield of whole-exome sequencing may offer advantages over traditional molecular diagnostic approaches in certain patients.”

Tammimies et al (2015) evaluated the molecular diagnostic yield of chromosomal microarray analysis (CMA) and whole-exome sequencing (WES) in children with spectrum disorder (ASD). The patient cohort included 258 consecutively enrolled unrelated children with ASD, stratified into three groups based on the presence of major congenital abnormalities and minor physical anomalies. All probands underwent CMA, with WES performed for 95 proband-parent trios. The molecular diagnostic yields of CMA and WES were comparable. Among the 95 patients undergoing WES, 8 children (8.4%) received an ASD-related molecular diagnosis. Among the children who underwent both CMA and WES testing, the estimated proportion with an identifiable genetic etiology was 15.8%. The investigators concluded that “if replicated in additional populations, these findings may inform appropriate selection of molecular diagnostic testing for children affected by ASD.”

Taylor et al (2015) performed whole genome sequencing in 217 individuals across a broad spectrum of genetic disorders in whom previous screening had identified no pathogenic variants. Disease-causing variants were identified in 21% of cases, with the proportion increasing to 34% (23/68) for mendelian disorders and 57% (8/14) in family trios. The investigators concluded that the results “demonstrate the value of genome sequencing for routine clinical diagnosis but also highlight many outstanding challenges.”

Miller et al (2016) performed exome/whole genome sequencing to identify the genetic cause in patients with , in whom prior clinically driven genetic testing had been negative. Out of the 40 patients tests, associated mutations were identified in 15 patients (37.5%), involving 14 different genes. In 5 of the 15 positive cases, the (molecular diagnosis had immediate, actionable consequences in patient management. The investigators concluded that the study results show “the benefits of exome/whole genome sequencing to identify causal mutations in craniosynostosis cases for which routine clinical testing has yielded negative results.”

Soden et al (2014) performed diagnostic WGS and/or WES in parent-child trios for hundred families with 119 children with neurodevelopmental disorders. 45 percent of the families received molecular diagnoses of an established genetic disorder disorders (53 of 119 affected children). An accelerated sequencing modality, rapid WGS, yielded diagnoses in 73% of families with acutely ill children (11 of 15). In this study, WES proved to be less costly than continued conventional diagnostic testing of children with NDD in whom initial testing failed to yield a diagnosis. The investigators concluded that “sequencing of or of trios should become an early part of the diagnostic workup of NDD and that accelerated sequencing modalities be extended to patients with high-acuity illness”

Shrivastava et al (2014) conducted a retrospective cohort study on 78 children with neurodevelopmental disabilities and unrevealing workup prior to whole exome sequencing. The overall presumptive diagnostic testing rate was 41% (n = 32 of 78 patients). Results of WES affected patient management in all cases, most often related to reproductive planning (n=27). The investigators concluded that the high diagnostic yield of WES could lead to earlier diagnosis, impacting medical management, prognostication, and family planning.

Hulick (2017) stated that next-generation sequencing (NGS) “may be appropriate for diagnosing suspected genetic disorders when sequencing of a single gene is unlikely to provide a diagnosis. NGS may be useful in the following situations:

Page 3 of 9 An Independent Licensee of the Blue Cross and Blue Shield Association

Whole Genome Whole Exome Sequencing AHS – M2032 “Notification” • One of several potential genes may be responsible • Obvious candidate genes have been tested and found to be normal • The cost of NGS would be less than that of sequencing individual candidate genes sequentially

Hulick (2017) recommended that “exome sequencing or whole genome sequencing should be considered when a condition demonstrates high heritability in a family or is suspected to have a genetic basis, but the number of potential candidate genes is large, or responsible gene(s) are unknown.” The author further stated that “one of the most common medical indications for whole genome sequencing or whole exome sequencing is evaluation of severe or developmental delay believed to have a genetic etiology in a patient with a negative initial evaluation. In some cases, evaluation of an affected child and both parents ("trio sequencing") is performed – especially when the inheritance pattern is dominant and a de novo is suspected. The value of NGS in this setting has been illustrated in several studies, in which the likelihood of reaching a molecular diagnosis is on the order of 25 percent.”

In summary, there is growing evidence supporting the use of WGS and WES in clinical practice. Further studies are required that directly compared WES/WGS with alternative testing strategies in terms of the diagnostic yield for pathogenic variants associated with the phenotype being evaluated.

Applicable Federal Regulations

Genotyping is considered a laboratory developed test (LDT); developed, validated and performed by individual laboratories.

LDTs are regulated by the Centers for Medicare and Medicaid (CMS) as high-complexity tests under the Clinical Laboratory Improvement Amendments of 1988 (CLIA’88).

As an LDT, the U. S. Food and Drug Administration has not approved or cleared this test; however, FDA clearance or approval is not currently required for clinical use.

Table 1: Examples of Laboratories Offering Exome Sequencing as a Clinical Service

Page 4 of 9 An Independent Licensee of the Blue Cross and Blue Shield Association

Whole Genome Whole Exome Sequencing AHS – M2032 “Notification” Laboratory Laboratory Indications for Testing

Ambry Genetics, Aliso "The patient's clinical presentation is unclear/atypical disease and there are Viejo, CA multiple genetic conditions in the differential diagnosis."

GeneDx, Gaithersburg, "a patient with a diagnosis that suggests the involvement of one or more of MD many different genes, which would, if even available and sequenced individually, be prohibitively expensive'

Baylor College of "Used when a patient's medical history and physical exam findings strongly , Houston TX suggest that there is an underlying genetic etiology. In some cases, the patient may have had an extensive evaluation consisting of multiple genetic test, without identifying an etiology."

University of California "This test is intended for use in conjunction with the clinical presentation and Los Angeles Health other markers of disease progression for the management of patients with rare System genetic disorders."

EdgeBio, Gaithersburg, Recommended " In situations where there has been a diagnostic failure with no MD discernible path. In situations where there are currently no available tests to determine the status of a potential genetic disease. In situations with atypical findings indicative of multiple disease(s)."

Children's Mercy Provided as a service to families with children who have had an extensive Hospitals and Clinics, negative work-up for a genetic disease; also used to identify novel disease Kansas City, MO genes.

Emory Genetics "Indicated when there is a suspicion of a genetic etiology contributing to the Laboratory, Atlanta, GA proband's manifestations."

Guidelines and Recommendations

Practice Guidelines and Position Statements

American College of Medical Genetics (ACMG)

In 2012, the ACMG released a policy statement outlining points to consider in the clinical application of genomic sequencing to the detection of germ-line mutations (ACMG, 2012). The ACMG recommended that WGS/WES should be considered in the clinical diagnostic assessment of a phenotypically affected individual when:

• “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.” • “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.” • “A patient presents with a likely genetic disorder, but specific genetic tests available for that phenotype have failed to arrive at a diagnosis.” Page 5 of 9 An Independent Licensee of the Blue Cross and Blue Shield Association

Whole Genome Whole Exome Sequencing AHS – M2032 “Notification” • “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 stated that “WGS/WES may be considered in preconception carrier screening, using a strategy to focus on genetic variants known to be associated with significant in homozygous or hemizygous progeny.” ACMG further stated that WGS and WES should not be used at this time as an approach to prenatal screening or as a first-tier approach for .

In 2013, ACMG published recommendations for reporting of incidental findings in clinical exome and genome sequencing. The ACMG recommended that when a report is issued for clinically indicated exome and genome sequencing, a minimum list of conditions, genes and variants should be evaluated and reported to the ordering clinician regardless of the indication for which the clinical sequencing was ordered.

In 2014 the ACMG published guidelines (Alford et al., 2014) for the clinical evaluation and etiologic diagnosis of hearing loss which state: “Pretest 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, whole- exome sequencing(WES), whole-genome sequencing (WGS), analysis, or microarray-based copy-number analysis, depending on clinical findings”.

American College of Obstetricians and Gynecologists (ACOG)

The ACOG (2016)published a committee opinion on Microarrays and Next-Generation Sequencing Technology: The Use of Advanced Genetic Diagnostic Tools in and Gynecology which states: “the routine use of whole-genome or whole-exome sequencing for prenatal diagnosis is not recommended outside of the context of clinical trials until sufficient peer reviewed data and validation studies are published.”

American Academy of (AAN)/American Association of Neuromuscular and Electrodiagnostic Medicine (AANEM)

The AAN/AANEM published guidelines (Kang et al., 2015) on the evaluation, diagnosis, and management of congenital muscular dystrophy which state: “In individuals with CMD who either do not have a mutation identified in one of the commonly associated genes or have a phenotype whose genetic origins have not been well characterized, might order whole-exome or whole-genome sequencing when those technologies become more accessible and affordable for routine clinical use (Level C).”

The AAN/AANEM published guidelines (Narayanaswami et al., 2014)on the diagnosis and treatment of limb-girdle and distal dystrophies which state: “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 (Level C).”

Center for Medicare and Medicaid Services

A. The CMS issued a national coverage decision (2018) which determined that next generation sequencing as a diagnostic laboratory test is reasonable and necessary, and covered nationally, when performed in a CLIA-certified laboratory, when ordered by a treating physician and when all of the following requirements are met:

Page 6 of 9 An Independent Licensee of the Blue Cross and Blue Shield Association

Whole Genome Whole Exome Sequencing AHS – M2032 “Notification”

1. Patient has: a. either recurrent, relapsed, refractory, metastatic, or advanced stages III or IV ; and b. either not been previously tested using the same NGS test for the same primary diagnosis of cancer or repeat testing using the same NGS test only when a new primary cancer diagnosis is made by the treating physician; and c. decided to seek further cancer treatment (e.g., therapeutic chemotherapy).

2. The diagnostic laboratory test using NGS must have: a. FDA approval or clearance as a companion in vitro diagnostic; and b. an FDA approved or cleared indication for use in that patient’s cancer; and c. results provided to the treating physician for management of the patient using a report template to specify treatment options.

B. Other Medicare Administrative Contractors (MACs) may determine coverage of other Next Generation Sequencing (NGS) as a diagnostic laboratory test for patients with cancer only when the test is performed in a CLIA-certified laboratory, ordered by a treating physician and the patient has: a. either recurrent, relapsed, refractory, metastatic, or advanced stages III or IV cancer; and b. either not been previously tested using the same NGS test for the same primary diagnosis of cancer or repeat testing using the same NGS test only when a new primary cancer diagnosis is made by the treating physician; and c. decided to seek further cancer treatment (e.g., therapeutic chemotherapy).

Billing/Coding/Physician Documentation Information This policy may apply to the following codes. Inclusion of a code in this section does not guarantee that it will be reimbursed. For further information on reimbursement guidelines, please see Administrative Policies on the Blue Cross Blue Shield of North Carolina web site at www.bcbsnc.com. They are listed in the Category Search on the Medical Policy search page.

Applicable service codes: 81415, 81416, 81417, 81425, 81426, 81427

Code Number PA Required PA Not Required Not Covered 81415 X 81416 X 81417 X 81425 X 81426 X 81427 X

BCBSNC may request medical records for determination of medical necessity. When medical records are requested, letters of support and/or explanation are often useful, but are not sufficient documentation unless all specific information needed to make a medical necessity determination is included.

Scientific Background and Reference Sources

Page 7 of 9 An Independent Licensee of the Blue Cross and Blue Shield Association

Whole Genome Whole Exome Sequencing AHS – M2032 “Notification” ACOG. (2016). Committee Opinion No.682: Microarrays and Next-Generation Sequencing Technology: The Use of Advanced Genetic Diagnostic Tools in Obstetrics and Gynecology. Obstet Gynecol, 128(6), e262-e268. doi:10.1097/aog.0000000000001817

Alford, R. L., Arnos, K. S., Fox, M., Lin, J. W., Palmer, C. G., Pandya, A., Yoshinaga-Itano, C. (2014). American College of Medical Genetics and guideline for the clinical evaluation and etiologic diagnosis of hearing loss. Genet Med, 16(4), 347-355. doi:10.1038/gim.2014.2

American College of Medical Genetics and Genomics (2012). Points to consider in the clinical application of genomic sequencing. Genetics in Medicine, 14 :759-761.

CMS. (2018). Decision Memo for Next Generation Sequencing (NGS) for Medicare Beneficiaries with Advanced Cancer (CAG-00450N). https://www.cms.gov/medicare-coverage- database/details/nca-decision-memo.aspx?NCAId=290

Hulick, P. (2014). The new era of whole exome sequencing. Retrieved April 4, 2017, from http://www.mdedge.com/internalmedicinenews/article/56906/gi-/new-era-whole-exome- sequencing

Lee, H., Deignan, J. L., Dorrani, N., Strom, S. P., Kantarci, S., Quintero-Rivera, F., Nelson, S. F. (2014). Clinical Exome Sequencing for Genetic Identification of Rare Mendelian Disorders. JAMA , 312 (18), 1880–1887. http://doi.org/10.1001/jama.2014.14604

Kang, P. B., Morrison, L., Iannaccone, S. T., Graham, R. J., Bonnemann, C. G., Rutkowski, A., . . . Griggs, R. C. (2015). Evidence-based guideline summary: evaluation, diagnosis, and management of congenital muscular dystrophy: 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, 84 (13), 1369-1378. doi:10.1212/wnl.0000000000001416

Miller, K.A., Twigg, S.R.F., McGowan, S.J., et al (2016) Diagnostic value of exome and whole genome sequencing in craniosynostosis. Journal of Medical Genetics Published Online First: 24 November 2016. doi: 10.1136/jmedgenet-2016-104215

NIH Intramural Sequencing Center. (2015). Whole Exome Sequencing and Analysis. Retrieved April 4, 2017, from https://www.nisc.nih.gov/docs/FAQ_whole_exome.pdf Narayanaswami, P., Weiss, M., Selcen, D., David, W., Raynor, E., Carter, G., Amato, A. A. (2014). 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, 83 (16), 1453-1463. doi:10.1212/wnl.0000000000000892

Rabbani, B., Tekin, M., and Mahdieh, N. (2014). The promise of whole-exome sequencing in medical genetics. Retrieved April 4, 2017, from http://www.nature.com/jhg/journal/v59/n1/full/jhg2013114a.html

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

Soden, S. E., Saunders, C. J., Willig, L. K., Farrow, E. G., Smith, L. D., Petrikin, J. E., … Kingsmore, S. F. (2014). Effectiveness of exome and genome sequencing guided by acuity of illness for diagnosis of neurodevelopmental disorders. Translational Medicine , 6(265), 265ra168. http://doi.org/10.1126/scitranslmed.3010076

Page 8 of 9 An Independent Licensee of the Blue Cross and Blue Shield Association

Whole Genome Whole Exome Sequencing AHS – M2032 “Notification” Tammimies, K., Marshall, C.R., Walker, S., et al (2015). Molecular Diagnostic Yield of Chromosomal Microarray Analysis and Whole-Exome Sequencing in Children With Autism Spectrum Disorder. JAMA, 314 (9):895-903.

Taylor, J.C., Martin, H.C., Lise, S., et al (2015). Factors influencing success of clinical genome sequencing across a broad spectrum of disorders. Nat Genet., 47 (7):717-726.

Yang, Y., Muzny, D. M., Xia, F., Niu, Z., Person, R., Ding, Y., Eng, C. M. (2014). Molecular Findings Among Patients Referred for Clinical Whole-Exome Sequencing. JAMA , 312 (18), 1870–1879. http://doi.org/10.1001/jama.2014.14601

Policy Implementation/Update Information 1/1/2019 BCBSNC will provide coverage for whole exome sequencing when it is considered to be medically necessary because criteria and guidelines are met. Whole genome sequencing is considered investigational for all indications. BCBSNC does not provide coverage for investigational services or procedures. Medical Director review 1/1/2019. Policy noticed 1/1/2019 for effective date 4/1/2019. (jd)

Medical policy is not an authorization, certification, explanation of benefits or a contract. Benefits and eligibility are determined before medical guidelines and payment guidelines are applied. Benefits are determined by the group contract and subscriber certificate that is in effect at the time services are rendered. This document is solely provided for informational purposes only and is based on research of current medical literature and review of common medical practices in the treatment and diagnosis of disease. Medical practices and knowledge are constantly changing and BCBSNC reserves the right to review and revise its medical policies periodically.

Page 9 of 9 An Independent Licensee of the Blue Cross and Blue Shield Association