Description of Procedure or Service
Cancer is defined as the uncontrolled growth and spread of abnormal cells and is increasingly shown to be initiated, propagated, and maintained by somatic genetic events (Johnson et al., 2014). About 1,688,780 new cancer cases are expected to be diagnosed in 2017 with about 600,920 Americans expected to die of cancer (~1650 people per day) (Siegel, Miller, & Jemal, 2017)
Analyses of gene expression can be clinically useful for disease classification, diagnosis, prognosis, and tailoring treatment to underlying genetic determinants of pharmacologic response (Spira, 2017). Precision or personalized oncology refers to evidence-based, individualized medicine that uses information about a person's genes, proteins, and environment to deliver the right care to the right cancer patient at the right time and results in measurable improvements in outcomes and a reduction on health care costs (M. Kalia, 2013).
***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 physician.
Policy
BCBSNC will provide coverage for molecular panel testing of cancers to identify targeted therapy when it is determined to be medically necessary because the medical criteria and guidelines shown below are met.
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 Molecular Panel Testing of Cancers to Identify Targeted Therapy is covered
Molecular panel testing of cancers to identify targeted therapy is considered medically necessary for multiplex panels of up to 50 genes that analyze for a subset of 5 or more genes considered to be standard-of-care for use with a given diagnosis, as defined in nationally recognized clinical guidelines such as those of the National Cancer Comprehensive Network (NCCN) or the American Society of Clinical Oncology (ASCO), Current genes scientifically shown to be impactful in the care of solid organ and hematolymphoid tumors are indicated in the coding table below. If less than 5 genes panel testing is needed please consult individual policies.
The covered panels must fit the American Medical Association’s Current Procedural Terminology (CPT®) codes for panels comprised of 5 to 50 genes for solid organ neoplasms (CPT® 81445) or hematolymphoid neoplasms or disorders (CPT® 81450) as shown in the coding table below.
When Molecular Panel Testing of Cancers to Identify Targeted Therapy is not covered
Panels containing more than 50 genes (CPT® code 81455) are investigational.
Coding Table
Specific genes for solid organ tumors and hematolymphoid neoplasms based on current NCCN guidelines are shown in table below. See individual policies for staging of cancers in which testing is appropriate.
Tumor Type | Disease State | Genes |
---|---|---|
Solid Tumor | Bone CA (Ewing Sarcoma) | EWSR1-ERG, EWSR1-ETV1, EWSR1-ETV4, EWSR1-FEV, EWSR1-FL1, FUS-ERG, FUS-FEV, |
Breast CA | BRCA1, BRCA2, ERBB2, PTEN, TP53, CDH1, STK11, 21 gene expression pattern, recurrence score | |
Solid Tumor | Non small cell lung CA (nonsquamous) | ALK, EGFR, ERBB2, KRAS, , ROS1 |
Colon CA | BRAF, CEACAM5, KRAS, MLH1, MSH2, MSH6, NRAS, PMS2, APC, EPCAM, MLH1, MSH2, MSH6, PMS2, MUTYH | |
Melanoma | BRAF, KIT | |
Myelodysplastic Syndromes | ASXL1, EZH2, ETV6, RUNX1, SF3B1, TP53, GATA2, JAK2, MPL, CALR, PDGFRB, RUNX1, TRG, TRA, TRB, TRD | |
Neuroendocrine CA | MEN1, RET | |
Ovarian CA | ATM, BRCA1, BRCA2, BRIP1, CHEK2, PALB2, RAD51C, RAD51D, MLH1, MLH2, MSH6, PMS2 | |
Pancreatic CA | MLH1, MSH2, MSH6, PMS2 | |
Penile CA | MLH1, MSH2, MSH6, PMS2 | |
Prostate CA | Men with clinically localized disease may consider the use of tumor-based molecular assays | |
Rectal CA | BRAF, MLH1, MSH2, MSH6, PMS2, NRAS, KRAS | |
Soft Tissue Sarcoma | APC, ATIC-ALK, CARS-ALK, CLTC-ALK, RANBP2-ALK, TPM3-ALK, TPM4-ALK, BRAF, COL1A1-PDGFB, CSF1-COL6A3, ETV6-NTRK3, EWSR1-ATF1, EWSR1-CREB1, FUS-ATF1, EWSR1-DDIT3, FUS-DDIT3, EWSR1-ERG, EWSR1-ETV1, EWSR1-ETV4, EWSR1-FEV, EWSR1-FLI1, EWSR1-PATZ1, FUS-ERG, EWSR1-NR4A3, TAF15-NR4A3, TCF12-NR4A3, TFG-NR4A3, EWSR1-WT1, FUS-CREB3L1, FUS-CREB3L2, GLI1, TSPAN31, CDK4, HMGA2, MDM2, HEY1-NCOA2, KIT, MYOD1, NAB-STAT6, NF1, CDKN2A, EED, SUZ12, PAX3-FOXO1, PAX3-FOXO4, PAX7-FOXO1, PDGFRA, SDHB, SDHC, SDHD, SMARCB1, SS18-SSX1, SS18-SSX2, SS18-SSX4, WWTR1-CAMTA1, YAP1-TFE3 | |
Thyroid CA | Abnormal gene/gene expression profiles, CALCA, CEACAM5, CGA, PTH, RET, TG, TSHB | |
Testicular CA | MLH1, MSH2, MSH6, PMS2 | |
Uterine CA | MLH1, MSH2, MSH6, PMS2 | |
Hematolymphoid | ALL | ABL1, BCR-ABL1, ETV-RUNX1, IL3-IGH, KMT2A, TCF3-PBX1 |
AML | ASXL1, BCR-ABL1, CBFB-MYH11, CEBPA, DEK-NUP214, DNMT3A, FLT3, IDH1, IDH2 KIT, MLL, MLLT3-MLL, NPM1, PML-RARA, RPN1-EV11, RUNX1, RUNX1-RUNX1T1, TET2, TP53, WT1 | |
CML | ABL1, BCR-ABL1, DNTT, MPO | |
CLL | BTK, TP53, IGHV | |
Primary Cutaneous B-Cell Lymphoma | BCL2 | |
Extranodal NK/T-Cell Lymphoma, nasal type | TRG, TRA, TRB, TRD | |
Peripheral T-Cell Lymphomas | ALK | |
Primary Cutaneous CD30+ T-Cell Lymphoproliferative Disorders | DUSP22, TRG, TRB | |
T-Cell Large Granular Lymphocytic Leukemia | STAT3, STAT5B, TRG, TRA, TRB, TRD | |
T-Cell Prolymphocytic Leukemia | TRA-TRD/MTCP1, TRA-TRD/TCL1A, TRG, TRA, TRB, TRD | |
Waldenstroms Macroglobulinemia/LymMacroglobulinemia/Lymphoplasmacytic Lymphoma | MyD88 L265, CXCR4 |
Policy Guidelines
Advances in sequencing technology have facilitated the identification of crucial genetic alterations that drive cancer cell growth by constitutive activation of cell signaling/cell cycling pathways or by inactivation of critical negative regulators of these networks (Bos, 1989; Davies et al., 2002; Levine & Oren, 2009; Soda et al., 2007). Compared with protein biomarkers, cancer genetic markers are more reproducible and less subject to intrinsic and extrinsic stimuli (M. Kalia, 2013; Li, Kung, Mack, & Gandara, 2016). The use of this information in personalized medicine has changed the paradigms in oncology because it is now based on understanding molecular carcinogenesis, pharmacogenomics, and individual genetic differences that determine the response to chemotherapeutics (Grullich & von Kalle, 2012; Madhu Kalia, 2015; Nalejska, Maczynska, & Lewandowska, 2014).
Small molecule inhibitors and antibodies have been developed that target particular oncogenic drivers (Chapman et al., 2011; Druker et al., 2001; Shaw et al., 2013; Slamon et al., 2001; Zhou et al., 2011). These targeted agents may be equivalent or even inferior to standard therapy in an unselected population but frequently induce dramatic regression in tumors harboring the target, demonstrating the value of precision medicine (Flaherty et al., 2010; Karapetis et al., 2008; Mok et al., 2009). Many agents are also now demonstrating signs of efficacy, even in previously difficult to target pathways involving activated RAS, impaired p53, and loss of cyclin-dependent kinase regulation(Ascierto et al., 2013; Dickson et al., 2013; Janne et al., 2013; Lehmann et al., 2012). Therefore, the ability to identify potentially actionable genetic alterations is imperative to exploiting the molecular vulnerabilities of cancer. (Johnson et al., 2014)
Figure from: (Dietel et al., 2015) See Appendix 1.
Currently, a variety of molecular diagnostic platforms are available (Meador et al., 2014). The most common clinically used sequencing platforms assess a limited number of the most extensively validated mutations (hotspots)(Dias-Santagata et al., 2010; Halait et al., 2012; Lovly et al., 2012; Shaw et al., 2013). These range from polymerase chain reaction (PCR)-based assays of a single point mutation to more extensive PCR- or mass spectrometry-based platforms assessing multiple point mutations across several genes (SNaPshot or Sequenom) (Halait et al., 2012; Lovly et al., 2012). However, activating mutations at non-hotspot locations that confer sensitivity to approved therapies (Bahadoran et al., 2013; Dahlman et al., 2012) and other clinically relevant gene fusions are not detected with hotspot testing methods (Drilon et al., 2013). A proportion of patients may therefore be excluded from potentially effective therapeutics based on incomplete genetic profiling(Johnson et al., 2014).
Whole genome and whole exome sequencing (WGS/WES) are available, however practical considerations related to data analysis, cost, and delay have constrained the widespread use of WGS/WES in clinics (Ulahannan, Kovac, Mulholland, Cazier, & Tomlinson, 2013).
Targeted NGS (i.e. FoundationOne) sequences the entire coding region of a large number of preselected genes with clinical or preclinical relevance in cancer (Wagle et al., 2012). Although less comprehensive than WGS/WES, targeted NGS does provide a comprehensive analysis of known genes with potential therapeutic and prognostic importance, a quick turnaround time (2–3 weeks in this case), and a standardized analytics pipeline (Frampton et al., 2013; Johnson et al., 2014).
The clinical utility of targeted therapy was examined by Kamps et al (2017)—See Appendix 2
Harada et al (2017) found that in 132 cases selected by a tumor board for comprehensive next generation sequencing, Forty-six cases (34.8%) had driver mutations that were associated with an active targeted therapeutic agent, including BRAF, PIK3CA, IDH1, KRAS, and BRCA1. An additional 56 cases (42.4%) had driver mutations previously reported in some type of cancer. Twenty-two cases (16.7%) did not have any clinically significant mutations. Eight cases did not yield adequate DNA. 15 cases were considered for targeted therapy, 13 of which received targeted therapy. One patient experienced a near complete response. Seven of 13 had stable disease or a partial response.
This approach of determining therapy based on genetic abnormalities rather than tissue of origin is increasingly important, as indicated by drugs such as pembrolizumab being developed and approved based on molecular indications (MSI-high or MMR deficient) independent of anatomical site of cancer origin (Hulick, 2018). However, the clinical utility and cost effectiveness of multigene panels versus broader sequencing methods is still in need of further study.
Applicable Federal Regulations
The FDA has approved more than 50 companion diagnostic devices to detect mutations in 12 different genes for the targeted treatment of cancer. Methodologies include immunohistochemistry, real-time or multiplex PCR, FISH, and next generation sequencing. The FDA has also approved additional nucleic acid based tests for cancer, not specifically as companion diagnostics.
On June 22, 2017 the FDA approved the Oncomine™ Dx Target Test (Thermo Fisher Scientific) as a next generation sequencing (NGS) test to detect multiple gene mutations for lung cancer in a single test from a single tissue specimen. This test detects the presence of BRAF, ROS1, and EGFR gene mutations or alterations in tumor tissue of patients with NSCLC. This test can be used to select patients with NSCLC with the BRAF V600E mutation for treatment with the combination of dabrafenib and trametinib.
On June 30, 2017 the FDA approved the Praxis Extended RAS Panel as a qualitative in vitro diagnostic test using targeted high throughput parallel sequencing for the detection of 56 specific mutations in RAS genes [KRAS (exons 2, 3, and 4) and NRAS (exons 2, 3, and 4)] in DNA extracted from formalin-fixed, paraffin-embedded (FFPE) colorectal cancer (CRC) tissue samples. The Praxis™ Extended RAS Panel is indicated to aid in the identification of patients with colorectal cancer for treatment with Vectibix® (panitumumab) based on a no mutation detected test result. The test is intended to be used on the Illumina MiSeqDx® instrument.
In November 2017 the FDA approved the marketing of the MSK-IMPACT assay as a qualitative in vitro diagnostic test that uses targeted next generation sequencing of formalin-fixed paraffin-embedded tumor tissue matched with normal specimens from patients with solid malignant neoplasms to detect tumor gene alterations in a broad multi gene panel. The test is intended to provide information on somatic mutations (point mutations and small insertions and deletions) and microsatellite instability for use by qualified health care professionals in accordance with professional guidelines, and is not conclusive or prescriptive for labeled use of any specific therapeutic product. MSK-IMPACT is a single-site assay performed at Memorial Sloan Kettering Cancer Center.
On November 30, 2017 the FDA approved FoundationOne CDx™ (F1CDx) as a next generation sequencing based in vitro diagnostic device for detection of substitutions, insertion and deletion alterations (indels), and copy number alterations (CNAs) in 324 genes and select gene rearrangements, as well as genomic signatures including microsatellite instability (MSI) and tumor mutational burden (TMB) using DNA isolated from formalin-fixed paraffin embedded (FFPE) tumor tissue specimens. The test is intended as a companion diagnostic to identify patients who may benefit from treatment with the targeted therapies in accordance with the approved therapeutic product labeling. Additionally, F1CDx is intended to provide tumor mutation profiling to be used by qualified health care professionals in accordance with professional guidelines in oncology for patients with solid malignant neoplasms. The F1CDx assay is a single-site assay performed at Foundation Medicine, Inc.
Guidelines and Recommendations
Practice Guidelines and Position Statements
National Comprehensive Cancer Network (NCCN)
NCCN guidelines for hereditary forms of cancers state that multi-gene testing should be offered to patients and families in the context of professional genetic expertise for pre- and post-test counseling. NCCN recommends that “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.” The guidelines state that “there may be a role for multi-gene testing in individuals who have tested negative (indeterminate) for a single syndrome, but whose personal or family history remains suggestive of an inherited susceptibility.” NCCN further recommends that “multi-gene testing can include intermediate penetrant (moderate-risk) genes”, but cautions that “not all genes included on available multi-gene tests are necessarily clinically actionable”.
Center for Medical Technology Policy (CMTP): Green Park Collaborative
In 2015, the Green Park Collaborative recommended that panels containing from 5 to 50 genes should be covered when the following criteria are met:
- A subset of at least 5 constituent genes or variants is cited in the label of an FDA-approved companion diagnostic indicated for the treatment of the patient; OR
- A subset of at least 5 constituent genes or variants is recommended for decision-making for the underlying diagnosis in nationally recognized clinical guidelines, such as those of the National Cancer Comprehensive Network (NCCN), or the American Society of Clinical Oncology (ASCO) or other guidelines that meet the IOM criteria for clinical guidelines; 10 OR
- A subset of at least 5 constituent genes are designated as standard of care for the underlying condition by the molecular testing committees of at least 3 NCCN member institutions; OR
- The provider has submitted two peer-reviewed journal articles of studies designed to demonstrate the safety and effectiveness of using the genomic information in question for clinical management of the patient’s diagnosis and support the conclusion that use of the information is reasonably likely to provide a health benefit for the patient.
AND, in all cases:
- The cost of analysis by NGS does not exceed the cost of individual sequencing of the target genes by other methods, AND
- The laboratory conducting the analysis is CLIA-certified and accredited by CAP for NGS testing.
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: 81170, 81206, 81445, 81450, 81455, 81479, 81599, 84999
Code Number | PPA Required | PPA not Required | Not Covered |
---|---|---|---|
81170 | X | ||
81206 | X | ||
81445 | X | ||
81450 | X | ||
81455 | X | ||
81479 | 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
Ascierto, P. A., Schadendorf, D., Berking, C., Agarwala, S. S., van Herpen, C. M., Queirolo, P., . . . Dummer, R. (2013). MEK162 for patients with advanced melanoma harbouring NRAS or Val600 BRAF mutations: a non-randomised, open-label phase 2 study. Lancet Oncol, 14(3), 249-256. doi:10.1016/s1470-2045(13)70024-x
Bahadoran, P., Allegra, M., Le Duff, F., Long-Mira, E., Hofman, P., Giacchero, D., . . . Ballotti, R. (2013). Major clinical response to a BRAF inhibitor in a patient with a BRAF L597R-mutated melanoma. J Clin Oncol, 31(19), e324-326. doi:10.1200/jco.2012.46.1061
Bos, J. L. (1989). ras oncogenes in human cancer: a review. Cancer Res, 49(17), 4682-4689.
Chapman, P. B., Hauschild, A., Robert, C., Haanen, J. B., Ascierto, P., Larkin, J., . . . McArthur, G. A. (2011). Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med, 364(26), 2507-2516. doi:10.1056/NEJMoa1103782
Dahlman, K. B., Xia, J., Hutchinson, K., Ng, C., Hucks, D., Jia, P., . . . Pao, W. (2012). BRAF(L597) mutations in melanoma are associated with sensitivity to MEK inhibitors. Cancer Discov, 2(9), 791-797. doi:10.1158/2159-8290.cd-12-0097
Davies, H., Bignell, G. R., Cox, C., Stephens, P., Edkins, S., Clegg, S., . . . Futreal, P. A. (2002). Mutations of the BRAF gene in human cancer. Nature, 417(6892), 949-954. doi:10.1038/nature00766
Dias-Santagata, D., Akhavanfard, S., David, S. S., Vernovsky, K., Kuhlmann, G., Boisvert, S. L., . . . Iafrate, A. J. (2010). Rapid targeted mutational analysis of human tumours: a clinical platform to guide personalized cancer medicine. EMBO Mol Med, 2(5), 146-158. doi:10.1002/emmm.201000070
Dickson, M. A., Tap, W. D., Keohan, M. L., D'Angelo, S. P., Gounder, M. M., Antonescu, C. R., . . . Schwartz, G. K. (2013). Phase II trial of the CDK4 inhibitor PD0332991 in patients with advanced CDK4-amplified well-differentiated or dedifferentiated liposarcoma. J Clin Oncol, 31(16), 2024-2028. doi:10.1200/jco.2012.46.5476
Dietel, M., Johrens, K., Laffert, M. V., Hummel, M., Blaker, H., Pfitzner, B. M., . . . Anagnostopoulos, I. (2015). A 2015 update on predictive molecular pathology and its role in targeted cancer therapy: a review focusing on clinical relevance. Cancer Gene Ther, 22(9), 417-430. doi:10.1038/cgt.2015.39
Drilon, A., Wang, L., Hasanovic, A., Suehara, Y., Lipson, D., Stephens, P., . . . Rizvi, N. (2013). Response to Cabozantinib in patients with RET fusion-positive lung adenocarcinomas. Cancer Discov, 3(6), 630-635. doi:10.1158/2159-8290.cd-13-0035
Druker, B. J., Talpaz, M., Resta, D. J., Peng, B., Buchdunger, E., Ford, J. M., . . . Sawyers, C. L. (2001). Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia. N Engl J Med, 344(14), 1031-1037. doi:10.1056/nejm200104053441401
Flaherty, K. T., Puzanov, I., Kim, K. B., Ribas, A., McArthur, G. A., Sosman, J. A., . . . Chapman, P. B. (2010). Inhibition of mutated, activated BRAF in metastatic melanoma. N Engl J Med, 363(9), 809-819. doi:10.1056/NEJMoa1002011
Frampton, G. M., Fichtenholtz, A., Otto, G. A., Wang, K., Downing, S. R., He, J., . . . Yelensky, R. (2013). Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat Biotechnol, 31(11), 1023-1031. doi:10.1038/nbt.2696
Grullich, C., & von Kalle, C. (2012). Recent developments and future perspectives of personalized oncology. Onkologie, 35 Suppl 1, 4-7. doi:10.1159/000334825
Halait, H., Demartin, K., Shah, S., Soviero, S., Langland, R., Cheng, S., . . . Lawrence, H. J. (2012). Analytical performance of a real-time PCR-based assay for V600 mutations in the BRAF gene, used as the companion diagnostic test for the novel BRAF inhibitor vemurafenib in metastatic melanoma. Diagn Mol Pathol, 21(1), 1-8. doi:10.1097/PDM.0b013e31823b216f
Harada, S., Arend, R., Dai, Q., Levesque, J. A., Winokur, T. S., Guo, R., . . . Yang, E. S. (2017). Implementation and utilization of the molecular tumor board to guide precision medicine. Oncotarget, 8(34), 57845-57854. doi:10.18632/oncotarget.18471
Hulick, P. (2018). Next-generation DNA sequencing (NGS): Principles and clinical applications - UpToDate. In J. Tirnauer (Ed.), UpToDate. Waltham, MA: UpToDate, Inc. Retrieved from https://www.uptodate.com/contents/next-generation-dna-sequencing-ngs-principles-and-clinical-applications?search=foundation%20one%20testing&source=search_result&selectedTitle=2~150&usage_type=default&display_rank=2#H3156709146.
Janne, P. A., Shaw, A. T., Pereira, J. R., Jeannin, G., Vansteenkiste, J., Barrios, C., . . . Crino, L. (2013). Selumetinib plus docetaxel for KRAS-mutant advanced non-small-cell lung cancer: a randomised, multicentre, placebo-controlled, phase 2 study. Lancet Oncol, 14(1), 38-47. doi:10.1016/s1470-2045(12)70489-8
Johnson, D. B., Dahlman, K. H., Knol, J., Gilbert, J., Puzanov, I., Means-Powell, J., . . . Pao, W. (2014). Enabling a Genetically Informed Approach to Cancer Medicine: A Retrospective Evaluation of the Impact of Comprehensive Tumor Profiling Using a Targeted Next-Generation Sequencing Panel. Oncologist, 19(6), 616-622. doi:10.1634/theoncologist.2014-0011
Kalia, M. (2013). Personalized oncology: recent advances and future challenges. Metabolism, 62 Suppl 1, S11-14. doi:10.1016/j.metabol.2012.08.016
Kalia, M. (2015). Biomarkers for personalized oncology: recent advances and future challenges. 64(3), S16–S21. doi:10.1016/j.metabol.2014.10.027
Kamps, R., Brandão, R. D., van den Bosch, B. J., Paulussen, A. D. C., Xanthoulea, S., Blok, M. J., & Romano, A. (2017). Next-Generation Sequencing in Oncology: Genetic Diagnosis, Risk Prediction and Cancer Classification. Int J Mol Sci, 18(2). doi:10.3390/ijms18020308
Karapetis, C. S., Khambata-Ford, S., Jonker, D. J., O'Callaghan, C. J., Tu, D., Tebbutt, N. C., . . . Zalcberg, J. R. (2008). K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N Engl J Med, 359(17), 1757-1765. doi:10.1056/NEJMoa0804385
Lehmann, S., Bykov, V. J., Ali, D., Andren, O., Cherif, H., Tidefelt, U., . . . Andersson, P. O. (2012). Targeting p53 in vivo: a first-in-human study with p53-targeting compound APR-246 in refractory hematologic malignancies and prostate cancer. J Clin Oncol, 30(29), 3633-3639. doi:10.1200/jco.2011.40.7783
Levine, A. J., & Oren, M. (2009). The first 30 years of p53: growing ever more complex. In Nat Rev Cancer (Vol. 9, pp. 749-758). England.
Li, T., Kung, H.-J., Mack, P. C., & Gandara, D. R. (2016). Genotyping and Genomic Profiling of Non–Small-Cell Lung Cancer: Implications for Current and Future Therapies. http://dx.doi.org/10.1200/JCO.2012.45.3753. doi:10.1200/JCO.2012.45.3753
Lovly, C. M., Dahlman, K. B., Fohn, L. E., Su, Z., Dias-Santagata, D., Hicks, D. J., . . . Pao, W. (2012). Routine multiplex mutational profiling of melanomas enables enrollment in genotype-driven therapeutic trials. PLoS One, 7(4), e35309. doi:10.1371/journal.pone.0035309
Meador, C. B., Micheel, C. M., Levy, M. A., Lovly, C. M., Horn, L., Warner, J. L., . . . Pao, W. (2014). Beyond histology: translating tumor genotypes into clinically effective targeted therapies. Clin Cancer Res, 20(9), 2264-2275. doi:10.1158/1078-0432.ccr-13-1591
Mok, T. S., Wu, Y. L., Thongprasert, S., Yang, C. H., Chu, D. T., Saijo, N., . . . Fukuoka, M. (2009). Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med, 361(10), 947-957. doi:10.1056/NEJMoa0810699
Nalejska, E., Maczynska, E., & Lewandowska, M. A. (2014). Prognostic and predictive biomarkers: tools in personalized oncology. Mol Diagn Ther, 18(3), 273-284. doi:10.1007/s40291-013-0077-9
Shaw, A. T., Kim, D. W., Nakagawa, K., Seto, T., Crino, L., Ahn, M. J., . . . Janne, P. A. (2013). Crizotinib versus chemotherapy in advanced ALK-positive lung cancer. N Engl J Med, 368(25), 2385-2394. doi:10.1056/NEJMoa1214886
Siegel, R. L., Miller, K. D., & Jemal, A. (2017). Cancer Statistics, 2017. CA Cancer J Clin, 67(1), 7-30. doi:10.3322/caac.21387
Slamon, D. J., Leyland-Jones, B., Shak, S., Fuchs, H., Paton, V., Bajamonde, A., . . . Norton, L. (2001). Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med, 344(11), 783-792. doi:10.1056/nejm200103153441101
Soda, M., Choi, Y. L., Enomoto, M., Takada, S., Yamashita, Y., Ishikawa, S., . . . Mano, H. (2007). Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature, 448(7153), 561-566. doi:10.1038/nature05945
Spira, A. S., Katrina. (2017). Tools for genetics and genomics: Gene expression profiling - UpToDate. In B. Raby (Ed.), UpToDate. Waltham. MA. Retrieved from https://www.uptodate.com/contents/tools-for-genetics-and-genomics-gene-expression-profiling?source=search_result&search=gene%20expression%20array%20cancer&selectedTitle=2~150.
Ulahannan, D., Kovac, M. B., Mulholland, P. J., Cazier, J. B., & Tomlinson, I. (2013). Technical and implementation issues in using next-generation sequencing of cancers in clinical practice. Br J Cancer, 109(4), 827-835. doi:10.1038/bjc.2013.416
Wagle, N., Berger, M. F., Davis, M. J., Blumenstiel, B., Defelice, M., Pochanard, P., . . . Garraway, L. A. (2012). High-throughput detection of actionable genomic alterations in clinical tumor samples by targeted, massively parallel sequencing. Cancer Discov, 2(1), 82-93. doi:10.1158/2159-8290.cd-11-0184
Zhou, C., Wu, Y. L., Chen, G., Feng, J., Liu, X. Q., Wang, C., . . . You, C. (2011). Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer (OPTIMAL, CTONG-0802): a multicentre, open-label, randomised, phase 3 study. Lancet Oncol, 12(8), 735-742. doi:10.1016/s1470-2045(11)70184-x
Policy Implementation/Update Information
1/1/2019 New policy developed. BCBSNC will provide coverage for molecular panel testing of cancers to identify targeted therapy when it is determined to be medically necessary and criteria are met. Medical Director review 1/1/2019. Policy noticed 1/1/2019 for effective date 4/1/2019. (lpr)
Appendix 1. Figure from: (Dietel et al., 2015)
Affected gene(s) | Type of alteration | Method for detection | Related treatment |
---|---|---|---|
HER2 | Amplification | IHC, ISH, | Herceptin |
PIKCA | SNV | Sequencing | Reduced response to anti-HER2 treatment, in particular doubl blockade (Trastuzumab /Lapatinib) |
Gene expression assays (EndoPre-dict, OncotypeDX etc.) | mRNA levels | qRT-PCR | Prognostic assay (endokrine Tx vs Chemoendokrine Tx) |
RAS (KRAS, NRAS) | SNV | Sequencing | Cetuximab / Panitumumab |
KIT | SNV, indel | Sequencing | Imatinib, Sunitinib |
PDGFR | SNV | Sequencing | Imatinib, Sunitinib |
BRAF | SNV | Sequencing | Vemurafenib, Dabrafenib, Trametinib |
KIT | SNV, indel | Sequencing | Sunitinib, Dasatinib, Imatinib |
EGFR | SNV, MNV, indel | Sequencing | Gefitinib, Erlotinib, Afatinib, Dacomitinib |
ALK | Translocation | ISH, IHC, Sequencing, | Crizotinib, Ceritinib, Alectinib |
ROS1 | Translocation | ISH, Sequencing | Crizotinib |
MET | Amplification | ISH, Sequencing | Resistance to EGFR TKIs |
BRCA1/BRCA2 | SNV, MNV, indel | Sequencing | Olaparib |
MYD88 | Mutation L265P and others | Sequencing | IRAK174 inhibitors |
CXCR4 | Mutation truncating C-terminal region | Sequencing | CXCR4 inhibitors,(plerixafor), blocking antibodies |
BRAF | V600E mutation | Sequencing | Vemurafenib |
MYC | Translocation, amplification, overexpression | IHC for MYC protein, FISH | BET inhibitors, Protein translation inhibitors |
Appendix 2. The clinical utility of targeted therapy was examined by Kamps et al (2017).
Genomic approach | Mean number of cancer-relevant somatic mutations (range) | Number of patients with tier 1 drug recommendations | Number of patients with tier 2 drug recommendations | Number of patients with actionable alterations | Mean number of actionable alterations (range) |
---|---|---|---|---|---|
Ion AmpliSeq Cancer Hotspot Panel v2 | 1.3 (0-4) | 24 (52 %) | 16 (35 %) | 24 (52 %) | 0.65 (0–3) |
Oncomine Comprehensive Panel | 2.5(0-11) | 39 (85 %) | 24 (52 %) | 41 (89 %) | 2.4 (0–6) |
FoundationOne | 3.7(0-22) | 39 (85 %) | 24 (52 %) | 41 (89 %) | 2.6 (0–7) |
This study | 17.3 (1-79) | 40 (87 %) | 26 (57 %) | 42 (91 %) | 4.9 (0–14) |