Section: Genetic Testing Last Reviewed Date: October 2015. Policy No: 73 Effective Date: November 1, 2015



Similar documents
Test Information Sheet

Molecular Pathology/Molecular Diagnostics/Genetic Testing

Test Information Sheet

TEST AND PRICE LIST FOR NEXT GENERATION SEQUENCING

1199 SEIU Benefit Funds Laboratory Management Program PRIOR AUTHORIZATION CODE LIST FOR OUTPATIENT MOLECULAR AND GENOMIC LABORATORY TESTS

JOHNS HOPKINS HEALTHCARE

Hereditary Breast Cancer Panels. High Risk Hereditary Breast Cancer Panel Hereditary Breast/Ovarian/Endometrial Cancer Panel

MEDICAL POLICY No R10 GENETICS: COUNSELING, TESTING, SCREENING

Common Cancers & Hereditary Syndromes

The Genetics of Early- Onset Breast Cancer. Cecelia Bellcross, Ph.D., M.S.,C.G.C. Department of Human Genetics Emory University School of Medicine

CPT Generic Test Test Description

6/10/2015. Hereditary Predisposition for Breast Cancer: Looking at BRCA1/BRCA2 Testing & Beyond. Hereditary Cancers. BRCA1 and BRCA2 Review

Hereditary Breast Cancer Testing. Diagnostic

Genetic Testing for Lynch Syndrome/Colorectal Cancer and Polyposis Syndromes

Ovarian Cancer Genetic Testing: Why, When, How?

Launching a Cancer Genetic Laboratory to Enhance Diagnosis and Treatment

Overview of testing for Lynch syndrome/hnpcc

Genetic Tests and Disease States Included in Humana s Genetic Guidance Program

NEW YORK STATE MEDICAID PROGRAM LABORATORY PROCEDURE CODES

Medical Policy Manual. Topic: Genetic Testing for Hereditary Breast and/or Ovarian Cancer. Date of Origin: January 27, 2011

PROVIDER POLICIES & PROCEDURES

GENETIC TESTING FOR INHERITED MUTATIONS OR SUSCEPTIBILITY TO CANCER OR OTHER CONDITIONS MED

Number Effective Date August 11, 2015 Revision Date(s) Replaces (not adopted)

Genetics and Breast Cancer. Elly Lynch, Senior Genetic Counsellor Manager, Austin Health Clinical Genetics Service

A Decision Support Tool to Facilitate Cancer Risk Assessment and Referral for Genetics Services. Kristen Vogel Postula, MS, CGC & Leigh Baumgart, PhD

Name of Policy: Genetic Testing for Inherited Cancer Predisposition and/or Pharmacogenetics related to Cancer Treatment

Genomic Medicine Education Initiatives of the College of American Pathologists

Genetic Testing for CHEK2 Mutations for Breast Cancer

Contents. molecular biology techniques. - Mutations in Factor II. - Mutations in MTHFR gene. - Breast cencer genes. - p53 and breast cancer

MEDICAL POLICY SUBJECT: GENETIC TESTING FOR HEREDITARY BRCA MUTATIONS. POLICY NUMBER: CATEGORY: Laboratory Test

Corporate Medical Policy Genetic Testing for Fanconi Anemia

Health First Health Plans Authorization List

National Medical Policy

What is Cancer? Cancer is a genetic disease: Cancer typically involves a change in gene expression/function:

Genetic Testing for Familial Adenomatous Polyposis and MYH-Associated Polyposis (Lynch Syndrome)

GENETIC CONSIDERATIONS IN CANCER TREATMENT AND SURVIVORSHIP

Genomic Medicine The Future of Cancer Care. Shayma Master Kazmi, M.D. Medical Oncology/Hematology Cancer Treatment Centers of America

Breast and Ovarian Cancer

Breast cancer and the role of low penetrance alleles: a focus on ATM gene

CLINICAL GUIDELINES. Lab Management Program. Effective January 15, 2016

IMMEDIATE HOT LINE: Effective March 2, 2015

MUTATION, DNA REPAIR AND CANCER

Molecular Diagnostics in Thyroid Cancer

Overview of the new AMA Molecular Pathology CPT codes

Public-Private Partnerships in early phase clinical research: Spurring access to innovative therapeutics

# Circuits relevant to cancer, related to known TFs.

targeted therapy a guide for the patient

patient guide BRCA1 and BRCA2 Genetic Testing for Hereditary Breast and Ovarian Cancer

La diagnostica molecolare nelle neoplasie colo-rettali. A do Scarpa. U iversita di Vero a

Beyond BRCA the Future is Now

Becker Muscular Dystrophy

Advice about familial aspects of breast cancer and epithelial ovarian cancer a guide for health professionals DECEMBER 2010

Hereditary Breast Cancer. Nicole Kounalakis, MD Assistant Professor of Surgery University of Colorado Medical Center

Cancer. 9p21.3 deletion. t(12;21) t(15;17)

NEIGE. diagnosis In oncogenetics. Nicolas Sévenet 02 juillet

Gene mutation and molecular medicine Chapter 15

Corporate Medical Policy Carrier Testing for Genetic Disease

Epi procolon The Blood Test for Colorectal Cancer Screening

Oncology Insights Enabled by Knowledge Base-Guided Panel Design and the Seamless Workflow of the GeneReader NGS System

Médecine de précision médecine personnalisée en Oncologie. Fabien Calvo, Directeur Recherche et Innovation, INCa, Directeur ITMO Cancer, AVIESAN

LAB MANAGEMENT CRITERIA. 1199SEIU Funds. Effective July 16, 2015

Genetic Testing for Susceptibility to Breast and Ovarian Cancer (BRCA1 and BRCA 2)

Progress and Prospects in Ovarian Cancer Screening and Prevention

Genetic Testing for Hereditary Breast and Ovarian Cancer - BRCA1/2 ANALYSIS -

Nuovi Scenari in Oncologia. G. Zoppoli X-Files in Nutrizione Clinica e Artificiale, 08/06/2012

RMHP Prior Authorization List Effective October 1, 2015 rev 7/18/2016

Name of Policy: Genetic Testing for Hereditary Breast and/or Ovarian Cancer

Genes and Cancer. What are genes? Dominant vs. recessive genes

BRCA1 & BRCA2: Genetic testing for hereditary breast and ovarian cancer patient guide

BAP1 germline mutations A new Cutaneous Nevus Melanoma Syndrome. Thomas Wiesner

Risk stratification for colorectal cancer especially: the difference between sporadic disease and polyposis syndromes. Dr. med. Henrik Csaba Horváth

BRCA in Men. Mary B. Daly,M.D.,Ph.D. June 25, 2010

Applications of comprehensive clinical genomic analysis in solid tumors: obstacles and opportunities

Prior to April, 2013, the preferred provider for outpatient labs was East Side Clinical Laboratory.

CancerTREATMENT NGS+ NSCLC Summary Report Page 1 of 7 PATIENT SPECIMEN PHYSICIAN

Microsatellite Instability (MSI) A New Paradigm in Cancer Treatment. Lynch Syndrome OUTLINE. GI Molecular Pathology

Colon Cancer Syndromes. Robin B. Mendelsohn MD Memorial Sloan Kettering Cancer Center Department of Medicine Gastroenterology and Nutrition Service

Special report. Chronic Lymphocytic Leukemia (CLL) Genomic Biology 3020 April 20, 2006

Downstream Outcomes of New Molecular Diagnostics CPT Coding System Codes

The following chapter is called "Preimplantation Genetic Diagnosis (PGD)".

BRCA1 and BRCA2. BRCA1 and BRCA2 Clinician Guide KNOWING WHAT TO LOOK FOR KNOWING WHERE TO LOOK AND KNOWING WHAT IT MEANS

Overview of Genetic Testing and Screening

SICKLE CELL ANEMIA & THE HEMOGLOBIN GENE TEACHER S GUIDE

Molecular Genetic Testing in Public Health and Clinical Settings

Human Genome Organization: An Update. Genome Organization: An Update

The Human Genome Project

Corporate Medical Policy

Corporate Medical Policy Molecular Markers in Fine Needle Aspirates of the Thyroid

CANCER RESEARCH UK STRATIFIED MEDICINE PROGRAMME IAN WALKER PHD, MBA HEAD OF STRATIFIED MEDICINE

Clinical Policy Title: Genomic tests in sensorineural hearing loss

BRCA1 & BRCA2 GeneHealth UK

Carrier Screening For Genetic Diseases Preconception Consent (Female and/or Male Partner)

Nuevas tecnologías basadas en biomarcadores para oncología

MEDICAL POLICY Genetic Testing

Lecture 3: Mutations

Data Analysis for Ion Torrent Sequencing

Advances in RainDance Sequence Enrichment Technology and Applications in Cancer Research. March 17, 2011 Rendez-Vous Séquençage

Haematopoietic Chimerism Analysis after Allogeneic Stem Cell Transplantation

patient education Fact Sheet PFS007: BRCA1 and BRCA2 Mutations MARCH 2015

BRCA and Breast/Ovarian Cancer -- Analytic Validity Version

Transcription:

Medical Policy Manual Topic: Genetic Cancer Susceptibility Panels Using Next Generation Sequencing Date of Origin: July 2014 Section: Genetic Testing Last Reviewed Date: October 2015 Policy No: 73 Effective Date: November 1, 2015 IMPORTANT REMINDER Medical Policies are developed to provide guidance for members and providers regarding coverage in accordance with contract terms. Benefit determinations are based in all cases on the applicable contract language. To the extent there may be any conflict between the Medical Policy and contract language, the contract language takes precedence. PLEASE NOTE: Contracts exclude from coverage, among other things, services or procedures that are considered investigational or cosmetic. Providers may bill members for services or procedures that are considered investigational or cosmetic. Providers are encouraged to inform members before rendering such services that the members are likely to be financially responsible for the cost of these services. Genetic testing for cancer susceptibility may be approached by a focused method that involves testing for well-characterized mutations based on a clinical suspicion of which gene(s) may be the cause of the familial cancer. Panel testing involves testing for multiple mutations in multiple genes at one time. Several companies, including Ambry Genetics and GeneDx, offer genetic testing panels that use nextgeneration sequencing methods for hereditary cancers. Next generation sequencing refers to one of several methods that use massively parallel platforms to allow the sequencing of large stretches of DNA. Panel testing is potentially associated with greater efficiencies in the evaluation of genetic diseases; however, it may provide information on genetic mutations that are of unclear clinical significance or which would not lead to changes in patient management. Currently available panels do not include all genes associated with hereditary cancer syndromes. In addition, these panels do not test for variants (i.e. single nucleotide polymorphisms [SNPs]), which may be associated with a low, but increased cancer risk. Next Generation Sequencing Cancer Panels A list of the genes that are included in these panels is given in Tables 1 and 2, followed by a brief description of each gene. Table 1: Ambry Genetics Hereditary Cancer Panel Tests 1 GT73

Gene BRCAplus GYNplus BreastNext OvaNext ColoNext PancNext PGLNext RenalNext CancerNext Tested BRCA1 BRCA2 ATM BARD1 BRIP1 MRE11A NBN RAD50 RAD51C PALB2 STK11 CHEK2 PTEN TP53 CDH1 MUTYH MLH1 MSH2 MSH6 EPCAM PMS2 APC BMPR1A SMAD4 NF1 RAD51D CDK4 CDKN2A RET SDHA SDHAF2 SDHB SDHC SDHD TMEM127 VHL FH FLCN MET MITF TSC1 TSC2 GeneDx offers a number of comprehensive cancer panels that use next generation sequencing, summarized in Table 2. Table 2: GeneDx Hereditary Cancer Panel Tests Gene Tested Breast/Ovarian Cancer Panel Breast Cancer High Risk Panel Endometrial Cancer Panel Lynch/ Colorectal Cancer High Risk Panel Colorectal Cancer Panel Pancreatic Cancer Panel Comprehensiv e Cancer Panel BRCA1 BRCA2 ATM BARD1 BRIP1 MRE11A NBN RAD50 RAD51C PALB2 STK11 CHEK2 PTEN TP53 CDH1 MUTYH MLH1 MSH2 2 GT73

MSH6 EPCAM PMS2 APC BMPR1A SMAD4 RAD51D CDK4 CDKN2A VHL RCC2 FANCC AIN2 Mayo Clinic also offers a hereditary colon cancer multi-gene panel analysis, which includes the genes in the Ambry Genetics ColoNext, with the addition of two other low-risk genes (MLH3 and AIN2). The University of Washington offers the BROCA Cancer Risk Panel, which is a next generation sequencing panel that includes the following mutations: AKT1, APC, ATM, ATR, BAP1, BARD1, BMPR1A, BRCA1, BRCA2, BRIP1, CDH1, CDK4, CDKN2A, CHEK1, CHEK2, CTNNA1, FAM175A, GALNT12, GEN1, GREM1, HOB13, MEN1, MLH1, MRE11A, MSH2 (+EPCAM), MSH6, MUTYH, NBN, PALB2, PIK3CA, PPM1D, PMS2, POLD1, POLE, PRSS1, PTEN, RAD50, RAD51, RAD51C, RAD51D, RET, SDHB, SDHC, SDHD, SMAD4, STK11, TP53, TP53BP1, VHL, and RCC2. [1] The University of Washington also offers the ColoSeq gene panel, which includes 19 genes associated with Lynch syndrome (hereditary non-polyposis colorectal cancer, HNPCC), familial adenomatous polyposis (FAP), MUTYH-associated polyposis (MAP), hereditary diffuse gastric cancer (HDGC), Cowden syndrome, Li-Fraumeni syndrome, Peutz-Jeghers syndrome, Muir-Torre syndrome, Turcot syndrome, and Juvenile Polyposis syndrome: AKT1, APC, BMPR1A, CDH1, EPCAM, GALNT12, GREM1, MLH1, MSH2, MSH6, MUTYH, PIK3CA, PMS2, POLE, POLD1, PTEN, SMAD4, STK11, and TP53. [2] Myriad Genetics offers myrisk Hereditary Cancer Panel, a comprehensive 25-gene cancer panel, that encompasses the simultaneous analysis of a larger number of genes combined in a panel for the identification of mutations impacting inherited risks for eight cancers: breast, colorectal, ovarian, endometrial, gastric, pancreatic, melanoma and prostate. Myriad does not identify the specific mutations. Regulatory Status Clinical laboratories may develop and validate tests in-house ( home-brew ) and market them as a laboratory service; such tests must meet the general regulatory standards of the Clinical Laboratory Improvement Act (CLIA). The laboratory offering the service must be licensed by CLIA for highcomplexity testing. Ambry Genetics is CLIA licensed. MEDICAL POLICY CRITERIA The following genetic cancer panels that use next generation sequencing are considered investigational because the current scientific evidence is not yet sufficient to establish how test results from panels which include a broad number of genes may be used to direct treatment decisions and improve health outcomes associated with all components of the panels: 3 GT73

Test Name BRCAplus Breast Plus Panel Breast Cancer High Risk Panel BreastNext Breast/Ovarian Cancer Panel BreastTrue High Risk Panel BROCA Cancer Risk Panel CancerNext Cancer NextGen Sequencing (NGS) Panel CancerTREATMENT NGS+ Comprehensive IHC & FISH ColoNext Colorectal Cancer Panel ColoSeq Comprehensive Cancer Panel Endometrial Cancer Panel FoundationOne FoundationOne Heme GYNplus ICG100 Lynch/Colorectal Cancer High Risk Panel myrisk Hereditary Cancer Panel OncoPlex Multiplexed Gene Sequencing Panel OvaNext PancNext Pancreatic Cancer Panel PGLNext RenalNext Solid Tumor Genomic Sequencing Panel Solid Tumor Mutation Panel Next Generation Sequencing SymGene68 Next Generation Sequencing Cancer Panel Laboratory Quest Diagnostics GeneDx GeneDx Pathway Genomics University of Washington Prevention Genetics biotheranostics GeneDx University of Washington GeneDx GeneDx Foundation Medicine, Inc. Foundation Medicine, Inc. Intermountain Cancer Genomics GeneDx Myriad University of Washington GeneDx Penn Medicine ARUP CellNetrix POLICY GUIDELINES It is critical that the list of information below is submitted for review to determine if the policy criteria are met. If any of these items are not submitted, it could impact our review and decision outcome. 1. Name of the genetic test(s) or panel test 2. Name of the performing laboratory and/or genetic testing organization (more than one may be listed) 3. The exact gene(s) and/or mutations being tested 4. Relevant billing codes 5. Brief description of how the genetic test results will guide clinical decisions that would not otherwise be made in the absence testing? 6. Medical records related to this genetic test History and physical exam Conventional testing and outcomes 4 GT73

Conservative treatment provided, if any SCIENTIFIC EVIDENCE Genetic cancer susceptibility panels utilizing next generation sequencing are best evaluated in the framework of a diagnostic test, as the test provides diagnostic information that assists in treatment decisions. Validation of the clinical use of any diagnostic test focuses on 3 main principles: 1. The analytic validity of the test, which refers to the technical accuracy of the test in detecting a mutation that is present or in excluding a mutation that is absent; 2. The clinical validity of the test, which refers to the diagnostic performance of the test (sensitivity, specificity, positive and negative predictive values) in detecting clinical disease; and 3. The clinical utility of the test, i.e., how the results of the diagnostic test will be used to change management of the patient and whether these changes in management lead to clinically important improvements in health outcomes. For genetic panels that test for a broad number of mutations, some components of the panel may be indicated based on the patient s clinical presentation and/or family history, while other components may not be indicated. The impact of test results related to the non-indicated mutations must be well-defined and take into account the possibility that the information may cause harm by leading to additional unnecessary interventions that would not otherwise be considered based on the patient s clinical presentation and/or family history. Analytic Validity Nonrandomized Study In order to determine whether next generation sequencing would enable accurate identification of inherited mutations for breast and ovarian cancer, Walsh and colleagues developed a genomic assay to capture, sequence, and detect all mutations in 21 genes, (which included 19 of the genes on the BreastNext and OvaNext panels). [3] Constitutional genomic DNA from individuals with known inherited mutations, was hybridized to custom oligonucleotides and then sequenced. The analysis was carried out blindly as to the mutation in each sample. All single nucleotide substitutions, small insertions and deletions, and large duplications and deletions were detected. There were no false positive results. Clinical Validity The published literature provides no guidance for the assessment of the clinical validity of panel testing for cancer susceptibility with next generation sequencing. Although it may be possible to evaluate the clinical validity of sequencing of individual genes found on these panels, the clinical validity of next generation sequencing for cancer susceptibility panels, which include mutations associated with an unknown or variable cancer risk, are of uncertain clinical validity. Clinical Utility Identifying an individual with a genetic mutation that confers a high risk of developing cancer could lead to changes in clinical management and improved health outcomes. There are well-defined clinical guidelines on the management of patients who are identified as having a high-risk hereditary cancer 5 GT73

syndrome. Changes in clinical management could include modifications in cancer surveillance, specific risk-reducing measures (e.g., prophylactic surgery), and treatment guidance (e.g., avoidance of certain exposures). In addition, other at-risk family members could be identified. On the other hand, identifying mutations that have intermediate or low penetrance is of limited clinical utility. Clinical management guidelines for patients found to have one of these mutations are not welldefined. In addition, there is a potential for harm, in that the diagnosis of an intermediate- or low-risk mutation may lead to undue psychological stress and unnecessary prophylactic surgical intervention. A limited body of literature exists on the potential clinical utility of available next generation sequencing (NGS) cancer panels. Nonrandomized Studies In 2014, in an industry-sponsored study, Cragun et al. reported the prevalence of clinically significant mutations and variants of uncertain significance among patients who underwent ColoNext panel testing. [4] For the period included in the study (March 2012-March 2013), the ColoNext test included the MLH1, MSH2, MSH6, PMS2, EPCAM, BMPR1, SMAD4, STK11, APC, MUTYH, CHEK2, TP53, PTEN, and CDH1 genes; alterations were classified as follows: (1) pathogenic mutation; (2) variant, likely pathogenic; (3) variant, unknown significance; (4) variant, likely benign; (5) benign. Data was analyzed for 586 patients whose ColoNext testing results and associated clinical data were maintained in a database by Ambry Genetics. Sixty-one patients (10.4%) had genetic alterations consistent with pathogenic mutations or likely pathogenic variants; after 8 patients with only CHEK2 or one MUTYH mutation were removed, 42 patients (7.2%) were considered to have actionable mutations. One hundred eighteen patients (20.1%) had at least one variant of uncertain significance, including 14 patients who had at least one variant of uncertain significance in addition to a pathologic mutation. Of the 42 patients with a pathologic mutation, most (30 patients, or 71%) clearly met National Comprehensive Cancer Network guidelines for syndrome-based testing, screening, or diagnosis, based on the available clinical and family history. The authors note that, The reality remains that syndrome based testing would have been sufficient to identify the majority of patients with deleterious mutations. Consequently, the optimal and most cost-effective use of panel-based testing as a first-tier test vs. a second tier test (i.e. after syndromebased testing is negative), remains to be determined. Mauer et al. reported a single academic center s genetics program s experience with next generation sequencing panels for cancer susceptibility. [5] The authors conducted a retrospective review of the outcomes and clinical indications for the ordering of Ambry s next generation sequencing panels (BreastNext, OvaNext, ColoNext, and CancerNext) for patients seen for cancer genetics counseling from April 2012 to January 2013. Of 1,521 new patients seen for cancer genetics counseling, 1,233 (81.1%) had genetic testing. Sixty of these patients (4.9% of the total) had a next generation sequencing panel ordered, 54 of which were ordered as a second-tier test after single-gene testing was performed. Ten tests were cancelled due to out-of-pocket costs or previously identified mutations. Of the 50 tests obtained, 5 were found to have a deleterious result (10%; compared with 131 [10.6%] of the 1,233 single-gene tests ordered at the same center during the study time frame). The authors report that of the 50 completed tests, 30 (60%) did not affect management decisions, 15 (30%) introduced uncertainty regarding the patients cancer risks, and 5 (10%) directly influenced management decisions. 6 GT73

In 2014, Kurian et al. evaluated the information from a NGS panel of 42 cancer associated genes in women who had been previously referred for clinical BRCA1/2 testing after clinical evaluation of hereditary breast and ovarian cancer from 2002 to 2012. [6] The authors aimed to assess concordance of the results of the panel with prior clinical sequencing, the prevalence of potentially clinically actionable results, and the downstream effects on cancer screening and risk reduction. Potentially actionable results were defined as pathogenic variants that cause recognized hereditary caner syndromes or have a published association with a two-fold or greater relative risk of breast cancer compared to average risk women. In total, 198 women participated in the study. Of these, 174 had breast cancer and 57 carried 59 germline BRCA 1/2 mutations. Testing with the panel confirmed 57 of 59 of the pathogenic BRCA ½ mutations; of the two others, one was detected but reclassified as a VUS and the other was a large insertion that would not be picked up by NGS panel testing. Of the women who tested negative for BRCA 1/2 mutations (n=141), 16 had pathogenic mutations in other genes (11.4%). The affected genes were ATM (n=2), BLM (n=1), CDH1 (n=1), CDKN2A (n=1), MLH1 (n=1), MUTYH (n=5), NBN (n=2), PRSS1 (n=1), and SL4 (n=2). Eleven of these variants had been previously reported in the literature and five were novel. 80% of the women with pathogenic mutations in the non BRCA1/2 genes had a personal history of breast cancer. Overall, a total of 428 VUS were identified in 39 genes, among 175 patients. Six women with mutations in ATM, BLM, CDH1, NBN and SL4 were advised to consider annual breast MRIs because of an estimated doubling of breast cancer risk, and six with mutations in CDH1, MLH1 and MUTYH were advised to consider frequent colonoscopy and/or endoscopic gastroduodenoscopy (once every 1-2 years) due to estimated increases in gastrointestinal cancer risk. One patient with a MLH1 mutation consistent with Lynch syndrome underwent risk-reducing salpingo-oophorectomy and early colonoscopy which identified a tubular adenoma that was excised (she had previously undergone hysterectomy for endometrial carcinoma). Conclusion Genetic cancer susceptibility panels using next generation sequencing for breast cancer, ovarian cancer, colon cancer or multiple cancer types (e.g., BreastNext, OvaNext, ColoNext and CancerNext, respectively) include mutations associated with varying risk of developing cancer. Therefore, these panels are of limited utility in that they may identify a clinically actionable mutation/syndrome, but could also identify a mutation for which there are no well-established guidelines or actionable level of risk associated with it. In addition, high rates of variants of uncertain significance have been reported with the use of these panels. [7] Clinical Practice Guidelines American Society of Clinical Oncology In a 2010 policy statement update on genetic and genomic testing for cancer susceptibility, the American Society of Clinical Oncology (ASCO) stated that testing for high-penetrance mutations in appropriate populations has clinical utility in that they inform clinical decision making and facilitate the prevention or amelioration of adverse health outcomes, but that genetic testing for intermediate-penetrance mutations are of uncertain clinical utility because the cancer risk associated with the mutation is generally too small to form an appropriate basis for clinical decision making. [8] ASCO recommends that genetic tests with uncertain clinical utility (low-to-moderate penetrance mutations) be administered in the context of clinical trials. 7 GT73

National Comprehensive Cancer Network The National Comprehensive Cancer Network (NCCN) guidelines on genetic/familial high-risk assessment for breast and ovarian cancer (v2.2015) [9] state the following regarding multi-gene testing: Patients who have a personal or family history suggestive of a single inherited cancer syndrome are most appropriately managed by genetic testing for that specific syndrome. When more than one gene can explain an inherited cancer syndrome, then multi-gene testing may be more efficient and/or cost effective. There is also a role for multi-gene testing in individuals who have tested negative (indeterminate) for a single syndrome, but whose personal or family history remains strongly suggestive of an inherited susceptibility. As commercially available tests differ in the specific genes analyzed (as well as classification of variants and many other factors), choosing the specific laboratory and test panel is important. Multi-gene testing can include intermediate penetrant (moderate risk) genes. For many of these genes, there are limited data on the degree of cancer risk and there are no clear guidelines on risk management for carriers of mutations. Not all genes included on available multi-gene test are necessarily clinically actionable. As is the case with high-risk genes, it is possible that the risks associated with moderate-risk genes may not be entirely due to that gene alone, but may be influenced by gene/gene or gene/environment interactions. Therefore, it may be difficult to use a known mutation alone to assign risk for relatives. In many cases, the information from testing for moderate penetrance genes does not change risk management compared to that based on family history alone. There is an increased likelihood of finding variants of unknown significance when testing for mutations in multiple genes. It is for these and other reasons that multi-gene testing is ideally offered in the context of professional genetic expertise for pre- and post-test counseling. Summary Genetic cancer panels that use next generation technology include mutations for a wide variety of cancers. The mutations included in these panels are associated with varying levels of risk of developing cancer, and only some of the mutations are associated with well-defined cancer syndromes which have established clinical management guidelines. The major advantage of these genetic panels is the ability to analyze many genes simultaneously, potentially improving the breadth and efficiency of genetic workup. Disadvantages of the panels are lower accuracy compared to direct sequencing, and the uncertain impact of a large amount of ancillary information related to non-indicated test results. Test results related to non-indicated mutations may potentially cause harm by leading to additional unnecessary interventions that would not otherwise be considered based on the patient s clinical presentation and/or family history. Currently, there is insufficient evidence to establish the clinical utility of the genetic cancer panels listed in the Medical Policy Criteria; therefore, these tests are considered investigational. Additional research is needed to demonstrate the clinical utility of simultaneous testing for all components of each panel test. REFERENCES 1. University of Washington. BROCA -- Cancer Risk Panel. [cited 7/6/2015]; Available from: http://web.labmed.washington.edu/tests/genetics/broca#broca_gene_list 8 GT73

2. University of Washington. ColoSeq -- Lynch and Polyposis Syndrome Panel. [cited 7/6/2015]; Available from: http://tests.labmed.washington.edu/coloseq 3. Walsh, T, Lee, MK, Casadei, S, et al. Detection of inherited mutations for breast and ovarian cancer using genomic capture and massively parallel sequencing. Proc Natl Acad Sci U S A. 2010;107:12629-33. PMID: 20616022 4. Cragun, D, Radford, C, Dolinsky, JS, Caldwell, M, Chao, E, Pal, T. Panel-based testing for inherited colorectal cancer: a descriptive study of clinical testing performed by a US laboratory. Clinical genetics. 2014 Feb 9. PMID: 24506336 5. Mauer, CB, Pirzadeh-Miller, SM, Robinson, LD, Euhus, DM. The integration of next-generation sequencing panels in the clinical cancer genetics practice: an institutional experience. Genet Med. 2014;16:407-12. PMID: 24113346 6. Kurian, AW, Hare, EE, Mills, MA, et al. Clinical evaluation of a multiple-gene sequencing panel for hereditary cancer risk assessment. United States, 2014. p. 2001-9. 7. Ambry Genetics TM NGS Cancer Panels - Breast, Ovarian, Colon and Endometrial Cancers.. [cited 7/6/2015]; Available from: http://www.ambrygen.com/sites/default/files/pdfs/acmg%20shared%20slides.pdf 8. Robson, ME, Storm, CD, Weitzel, J, Wollins, DS, Offit, K. American Society of Clinical Oncology policy statement update: genetic and genomic testing for cancer susceptibility. J Clin Oncol. 2010;28:893-901. PMID: 20065170 9. National Comprehensive Cancer Network (NCCN). Clinical Practice Guidelines in Oncology TM. Genetic/Familial High-Risk Assessment: Breast and Ovarian v.2.2015. [cited 07/06/2015]; Available from: http://www.nccn.org/professionals/physician_gls/pdf/genetics_screening.pdf CROSS REFERENCES Genetic Testing for Hereditary Breast and/or Ovarian Cancer, Genetic Testing, Medical Policy No. 2 Genetic Testing for Inherited Susceptibility to Colon Cancer, Genetic Testing, Medical Policy No. 6 Evaluating the Utility of Genetic Panels, Genetic Testing, Medical Policy No. 64 CODES NUMBER There are no specific codes for molecular pathology testing by panels. If the specific analyte is listed in CPT codes 81200-81355 or 81400-81408, the specific CPT code would be reported. If the specific analyte is not listed in the more specific CPT codes, unlisted code 81479 would be reported. The unlisted code would be reported once to represent all of the unlisted analytes in the panel. CPT 81200 ASPA (aspartoacylase) (eg, Canavan disease) gene analysis, common variants (eg, E285A, Y231) 81201 APC (adenomatous polyposis coli) (eg, familial adenomatosis polyposis [FAP], attenuated FAP) gene analysis; full gene sequence 81202 ;known familial variants 81203 ;duplication/deletion variants 9 GT73

CODES NUMBER 81205 BCKDHB (branched-chain keto acid dehydrogenase E1, beta polypeptide) (eg, maple syrup urine disease) gene analysis, common variants (eg, R183P, G278S, E422) 81206 BCR/ABL1 (t(9;22)) (eg, chronic myelogenous leukemia) translocation analysis; major breakpoint, qualitative or quantitative 81207 ;minor breakpoint, qualitative or quantitative 81208 ;other breakpoint, qualitative or quantitative 81209 BLM (Bloom syndrome, RecQ helicase-like) (eg, Bloom syndrome) gene analysis, 2281del6ins7 variant 81210 BRAF (v-raf murine sarcoma viral oncogene homolog B1) (eg, colon cancer), gene analysis, V600E variant 81211 BRCA1, BRCA2 (breast cancer 1 and 2) (eg, hereditary breast and ovarian cancer) gene analysis; full sequence analysis and common duplication/deletion variants in BRCA1 (ie, exon 13 del 3.835kb, exon 13 dup 6kb, exon 14-20 del 26kb, exon 22 del 510bp, exon 8-9 del 7.1kb) 81212 ;185delAG, 5385insC, 6174delT variants 81213 ;uncommon duplication/deletion variants 81214 BRCA1 (breast cancer 1) (eg, hereditary breast and ovarian cancer) gene analysis; full sequence analysis and common duplication/deletion variants (ie, exon 13 del 3.835kb, exon 13 dup 6kb, exon 14-20 del 26kb, exon 22 del 510bp, exon 8-9 del 7.1kb) 81215 ;known familial variant 81216 BRCA2 (breast cancer 2) (eg, hereditary breast and ovarian cancer) gene analysis; full sequence analysis 81217 ;known familial variant 81220 CFTR (cystic fibrosis transmembrane conductance regulator) (eg, cystic fibrosis) gene analysis; common variants (eg, ACMG/ACOG guidelines) 81221 ;known familial variants 81222 ;duplication/deletion variants 81223 ;full gene sequence 81224 ;intron 8 poly-t analysis (eg, male infertility) 10 GT73

CODES NUMBER 81225 CYP2C19 (cytochrome P450, family 2, subfamily C, polypeptide 19) (eg, drug metabolism), gene analysis, common variants (eg, *2, *3, *4, *8, *17) 81226 CYP2D6 (cytochrome P450, family 2, subfamily D, polypeptide 6) (eg, drug metabolism), gene analysis, common variants (eg, *2, *3, *4, *5, *6, *9, *10, *17, *19, *29, *35, *41, *1N, *2N, *4N) 81227 CYP2C9 (cytochrome P450, family 2, subfamily C, polypeptide 9) (eg, drug metabolism), gene analysis, common variants (eg, *2, *3, *5, *6) 81228 Cytogenomic constitutional (genome-wide) microarray analysis; interrogation of genomic regions for copy number variants (eg, bacterial artificial chromosome [BAC] or oligo-based comparative genomic hybridization [CGH] microarray analysis) 81229 ;interrogation of genomic regions for copy number and single nucleotide polymorphism (SNP) variants for chromosomal abnormalities 81235 EGFR (epidermal growth factor receptor) (eg, non-small cell lung cancer) gene analysis, common variants (eg, exon 19 LREA deletion, L858R, T790M, G719A, G719S, L861Q) 81240 F2 (prothrombin, coagulation factor II) (eg, hereditary hypercoagulability) gene analysis, 20210G>A variant 81241 F5 (coagulation factor V) (eg, hereditary hypercoagulability) gene analysis, Leiden variant 81242 FANCC (Fanconi anemia, complementation group C) (eg, Fanconi anemia, type C) gene analysis, common variant (eg, IVS4+4A>T) 81243 FMR1 (fragile mental retardation 1) (eg, fragile mental retardation) gene analysis; evaluation to detect abnormal (eg, expanded) alleles 81244 ;characterization of alleles (eg, expanded size and methylation status) 81245 FLT3 (fms-related tyrosine kinase 3) (eg, acute myeloid leukemia), gene analysis; internal tandem duplication (ITD) variants (ie, exons 14, 15) 81246 ;tyrosine kinase domain (TKD) variants (eg, D835, I836) 81250 G6PC (glucose-6-phosphatase, catalytic subunit) (eg, Glycogen storage disease, type 1a, von Gierke disease) gene analysis, common variants (eg, R83C, Q347) 81251 GBA (glucosidase, beta, acid) (eg, Gaucher disease) gene analysis, common variants (eg, N370S, 84GG, L444P, IVS2+1G>A) 11 GT73

CODES NUMBER 81252 GJB2 (gap junction protein, beta 2, 26kDa, connexin 26) (eg, nonsyndromic hearing loss) gene analysis; full gene sequence 81253 ;known familial variants 81254 GJB6 (gap junction protein, beta 6, 30kDa, connexin 30) (eg, nonsyndromic hearing loss) gene analysis, common variants (eg, 309kb [del(gjb6- D13S1830)] and 232kb [del(gjb6-d13s1854)]) 81255 HEA (hexosaminidase A [alpha polypeptide]) (eg, Tay-Sachs disease) gene analysis, common variants (eg, 1278insTATC, 1421+1G>C, G269S) 81256 HFE (hemochromatosis) (eg, hereditary hemochromatosis) gene analysis, common variants (eg, C282Y, H63D) 81257 HBA1/HBA2 (alpha globin 1 and alpha globin 2) (eg, alpha thalassemia, Hb Bart hydrops fetalis syndrome, HbH disease), gene analysis, for common deletions or variant (eg, Southeast Asian, Thai, Filipino, Mediterranean, alpha3.7, alpha4.2, alpha20.5, and Constant Spring) 81260 IKBKAP (inhibitor of kappa light polypeptide gene enhancer in B-cells, kinase complex-associated protein) (eg, familial dysautonomia) gene analysis, common variants (eg, 2507+6T>C, R696P) 81261 IGH@ (Immunoglobulin heavy chain locus) (eg, leukemias and lymphomas, B-cell), gene rearrangement analysis to detect abnormal clonal population(s); amplified methodology (eg, polymerase chain reaction) 81262 ;direct probe methodology (eg, Southern blot) 81263 IGH@ (Immunoglobulin heavy chain locus) (eg, leukemia and lymphoma, B- cell), variable region somatic mutation analysis 81264 IGK@ (Immunoglobulin kappa light chain locus) (eg, leukemia and lymphoma, B-cell), gene rearrangement analysis, evaluation to detect abnormal clonal population(s) 81265 Comparative analysis using Short Tandem Repeat (STR) markers; patient and comparative specimen (eg, pre-transplant recipient and donor germline testing, post-transplant non-hematopoietic recipient germline [eg, buccal swab or other germline tissue sample] and donor testing, twin zygosity testing, or maternal cell contamination of fetal cells) 81266 ;each additional specimen (eg, additional cord blood donor, additional fetal samples from different cultures, or additional zygosity in multiple birth pregnancies) (List separately in addition to code for primary procedure) 12 GT73

CODES NUMBER 81267 Chimerism (engraftment) analysis, post transplantation specimen (eg, hematopoietic stem cell), includes comparison to previously performed baseline analyses; without cell selection 81268 ;with cell selection (eg, CD3, CD33), each cell type 81270 JAK2 (Janus kinase 2) (eg, myeloproliferative disorder) gene analysis, p.val617phe (V617F) variant 81275 KRAS (v-ki-ras2 Kirsten rat sarcoma viral oncogene) (eg, carcinoma) gene analysis, variants in codons 12 and 13 81280 Long QT syndrome gene analyses (eg, KCNQ1, KCNH2, SCN5A, KCNE1, KCNE2, KCNJ2, CACNA1C, CAV3, SCN4B, AKAP, SNTA1, and ANK2); full sequence analysis 81281 ;known familial sequence variant 81282 ;duplication/deletion variants 81287 MGMT (O-6-methylguanine-DNA methyltransferase) (eg, glioblastoma multiforme), methylation analysis 81288 MLH1 (mutl homolog 1, colon cancer, nonpolyposis type 2) (eg, hereditary non-polyposis colorectal cancer, Lynch syndrome) gene analysis; promoter methylation analysis 81290 MCOLN1 (mucolipin 1) (eg, Mucolipidosis, type IV) gene analysis, common variants (eg, IVS3-2A>G, del6.4kb) 81291 MTHFR (5,10-methylenetetrahydrofolate reductase) (eg, hereditary hypercoagulability) gene analysis, common variants (eg, 677T, 1298C) 81292 MLH1 (mutl homolog 1, colon cancer, nonpolyposis type 2) (eg, hereditary non-polyposis colorectal cancer, Lynch syndrome) gene analysis; full sequence analysis 81293 ;known familial variants 81294 ;duplication/deletion variants 81295 MSH2 (muts homolog 2, colon cancer, nonpolyposis type 1) (eg, hereditary non-polyposis colorectal cancer, Lynch syndrome) gene analysis; full sequence analysis 81296 ;known familial variants 81297 ;duplication/deletion variants 13 GT73

CODES NUMBER 81298 MSH6 (muts homolog 6 [E. coli]) (eg, hereditary non-polyposis colorectal cancer, Lynch syndrome) gene analysis; full sequence analysis 81299 ;known familial variants 81300 ;duplication/deletion variants 81301 Microsatellite instability analysis (eg, hereditary non-polyposis colorectal cancer, Lynch syndrome) of markers for mismatch repair deficiency (eg, BAT25, BAT26), includes comparison of neoplastic and normal tissue, if performed 81302 MECP2 (methyl CpG binding protein 2) (eg, Rett syndrome) gene analysis; full sequence analysis 81303 ;known familial variant 81304 ;duplication/deletion variants 81310 NPM1 (nucleophosmin) (eg, acute myeloid leukemia) gene analysis, exon 12 variants 81315 PML/RARalpha, (t(15;17)), (promyelocytic leukemia/retinoic acid receptor alpha) (eg, promyelocytic leukemia) translocation analysis; common breakpoints (eg, intron 3 and intron 6), qualitative or quantitative 81316 ;single breakpoint (eg, intron 3, intron 6 or exon 6), qualitative or quantitative 81317 PMS2 (postmeiotic segregation increased 2 [S. cerevisiae]) (eg, hereditary non-polyposis colorectal cancer, Lynch syndrome) gene analysis; full sequence analysis 81318 ;known familial variants 81319 ;duplication/deletion variants 81321 PTEN (phosphatase and tensin homolog) (eg, Cowden syndrome, PTEN hamartoma tumor syndrome) gene analysis; full sequence analysis 81322 ;known familial variant 81323 ;duplication/deletion variant 81324 PMP22 (peripheral myelin protein 22) (eg, Charcot-Marie-Tooth, hereditary neuropathy with liability to pressure palsies) gene analysis; duplication/deletion analysis 14 GT73

CODES NUMBER 81325 ;full sequence analysis 81326 ;known familial variant 81330 SMPD1(sphingomyelin phosphodiesterase 1, acid lysosomal) (eg, Niemann- Pick disease, Type A) gene analysis, common variants (eg, R496L, L302P, fsp330) 81331 SNRPN/UBE3A (small nuclear ribonucleoprotein polypeptide N and ubiquitin protein ligase E3A) (eg, Prader-Willi syndrome and/or Angelman syndrome), methylation analysis 81332 SERPINA1 (serpin peptidase inhibitor, clade A, alpha-1 antiproteinase, antitrypsin, member 1) (eg, alpha-1-antitrypsin deficiency), gene analysis, common variants (eg, *S and *Z) 81340 TRB@ (T cell antigen receptor, beta) (eg, leukemia and lymphoma), gene rearrangement analysis to detect abnormal clonal population(s); using amplification methodology (eg, polymerase chain reaction) 81341 ;using direct probe methodology (eg, Southern blot) 81342 TRG@ (T cell antigen receptor, gamma) (eg, leukemia and lymphoma), gene rearrangement analysis, evaluation to detect abnormal clonal population(s) 81350 UGT1A1 (UDP glucuronosyltransferase 1 family, polypeptide A1) (eg, irinotecan metabolism), gene analysis, common variants (eg, *28, *36, *37) 81355 VKORC1 (vitamin K epoxide reductase complex, subunit 1) (eg, warfarin metabolism), gene analysis, common variants (eg, -1639/3673) 81400 Molecular pathology procedure, Level 1 81401 Molecular pathology procedure, Level 2 81402 Molecular pathology procedure, Level 3 81403 Molecular pathology procedure, Level 4 81404 Molecular pathology procedure, Level 5 81405 Molecular pathology procedure, Level 6 81406 Molecular pathology procedure, Level 7 81407 Molecular pathology procedure, Level 8 81408 Molecular pathology procedure, Level 9 15 GT73

CODES NUMBER 81455 Targeted genomic sequence analysis panel, solid organ or hematolymphoid neoplasm, DNA and RNA analysis when performed, 51 or greater genes (eg, ALK, BRAF, CDKN2A, CEBPA, DNMT3A, EGFR, ERBB2, EZH2, FLT3, IDH1, IDH2, JAK2, KIT, KRAS, MLL, NPM1, NRAS, MET, NOTCH1, PDGFRA, PDGFRB, PGR, PIK3CA, PTEN, RET), interrogation for sequence variants and copy number variants or rearrangements, if performed 81479 Unlisted molecular pathology procedure HCPCS None 16 GT73