Coverage Policy Manual
Policy #: 2015002
Category: Laboratory
Initiated: January 2015
Last Review: November 2023
  Genetic Test: Somatic Biomarker testing (including Liquid Biopsy) for Targeted Treatment and Immunotherapy in Non-Small-Cell Lung Cancer (EGFR, ALK, BRAF, ROS1, RET, MET, KRAS, HER2, PD-L1, TMB)

Description:
Treatment options for non-small cell lung cancer (NSCLC) depend on disease stage and include various combinations of surgery, radiotherapy, systemic therapy, and best supportive care. Unfortunately, in up to 85% of cases, the cancer has spread locally beyond the lungs at diagnosis, precluding surgical eradication. In addition, up to 40% of patients with NSCLC present with metastatic disease (Fathi, 2008). When treated with standard platinum based chemotherapy, patients with advanced NSCLC have a median survival of 8 to 11 months and a 1-year survival of 30% to 45% (Martoni, 2005; Rudd, 2005). The identification of specific, targetable oncogenic “driver” mutations in a subset of NSCLCs has resulted in a reclassification of lung tumors to include molecular subtypes, which are predominantly of adenocarcinoma histology
 
EGFR
The epidermal growth factor receptor (EGFR), a receptor tyrosine kinase (TK), is frequently overexpressed and activated in non-small-cell lung cancer (NSCLC). Drugs that inhibit EGFR signaling either prevent ligand binding to the extracellular domain (monoclonal antibodies) or inhibit intracellular TK activity (small molecule tyrosine kinase inhibitors [TKIs]). These targeted therapies dampen signal transduction through pathways downstream to the EGF receptor, such as the RAS/RAF/MAPK cascade. RAS proteins are G-proteins that cycle between active and inactive forms in response to stimulation from cell surface receptors such as EGFR, acting as binary switches between cell surface EGFR and downstream signaling pathways. These pathways are important in cancer cell proliferation, invasion, metastasis, and stimulation of neovascularization.
 
Somatic variants in the tyrosine kinase domain of the EGFR gene, notably small deletions in exon 19 and a point mutation in exon 21 (L858R, indicating substitution of leucine by arginine at codon position 858) are the most commonly found EGFR variants associated with sensitivity to EGFR tyrosine kinase inhibitors (TKIs; afatinib, erlotinib, gefitinib). These variants are referred to as sensitizing variants. Almost all patients who initially respond to an EGFR TKI experience disease progression. The most common of these secondary variants, called resistance variants, involves the substitution of methionine for threonine at position 790 (T790M) on exon20.
 
Fang et al reported EGFR variants (all L858R) in 3 (2%) of 146 consecutively treated Chinese patients with early-stage squamous cell carcinoma (SCC) (Fang, 2013). In a separate cohort of 63 Chinese patients with SCC who received erlotinib or gefitinib as second-or third-line treatment (63% never-smokers, 21% women), EGFR variant prevalence (all exon 19 deletion or L858R) was 23.8%.
 
In a comprehensive analysis of 14 studies involving 2880 patients, Mitsudomi et al reported EGFR variants in 10% of men, 7%of non-Asian patients, 7% of current or former smokers, and 2% of patients with nonadenocarcinoma histologies (Mitsudomi, 2006). Eberhard et al, observed EGFR variants in 6.4% of patients with SCC and Rosell et al, observed EGFR variants in 11.5% of patients with large cell carcinomas (Eberhard, 2005; Rosell, 2009). Both studies had small sample sizes.
 
In 2 other studies, the acquired EGFR T790M variant has been estimated to be present in 50% to 60% of TKI-resistant cases in approximately 200 patients (Yu, 2013; Sequist, 2011).
 
ALK
ALK (anaplastic large cell lymphoma kinase) is a tyrosine kinase that, in NSCLC, is aberrantly activated because of a chromosomal rearrangement which leads to a fusion gene and expression of a protein with constitutive tyrosine kinase activity that has been demonstrated to play a role in controlling cell proliferation. The EML4-ALK fusion gene results from an inversion within the short arm of chromosome 2.
 
The EML4-ALK rearrangement (“ALK-positive”) is detected in 3% to 6% of NSCLC patients, with the highest prevalence in never-smokers or light ex-smokers who have adenocarcinoma.
  
BRAF
RAF proteins are serine/threonine kinases that are downstream of RAS in the RAS-RAF-ERK-MAPK pathway. In this pathway, the BRAF gene is the most frequently mutated in NSCLC, in 1% to 3% of adenocarcinomas. Unlike melanoma, about 50% of the variants in NSCLC are non-V600E variants (Thunnissen, 2014). Most BRAF variants occur more frequently in smokers.
 
ROS1
ROS1 codes for a receptor tyrosine kinase of the insulin receptor family, and chromosomal rearrangements result in fusion genes. The prevalence of ROS1 fusions in NSCLC varies from 0.9-3.7%. (Thunnissen, 2014). Patients with ROS1 fusions are typically never smokers with adenocarcinoma.
 
KRAS
The KRAS gene (which encodes RAS proteins) can harbor oncogenic mutations that result in a constitutively activated protein, independent of signaling from the EGF receptor, possibly rendering a tumor resistant to therapies that target the EGF receptor. Variants in the KRAS gene, mainly codons 12 and 13, have been reported in 20-30% of NSCLC, and occur most often in adenocarcinomas in heavy smokers.
 
KRAS variants can be detected by direct sequencing, PCR technologies, or NGS.
 
EGFR, ALK, ROS1, and KRAS driver mutations are considered to be mutually exclusive.
 
HER2
Her2 is a member of the HER (EGFR) family of TK receptors and has no specific ligand. When activated, it forms dimers with other EGFR family members. Her2 is expressed in approximately 25% of NSCLC. Her2 mutations are detected mainly in exon 20 in 1-2% of NSCLC, predominantly in adenocarcinomas in nonsmoking women (Thunnissen, 2014).
 
RET
RET (rearranged during transfection) is a proto-oncogene that encodes a receptor tyrosine kinase growth factor. Translocations that result in fusion genes with several partners have been reported.4 RET fusions occur in 0.6-2% of NSCLCs and in 1.2-2% of adenocarcinomas (Thunnissen, 2014).
 
MET
MET alteration is one of the critical events for acquired resistance in EGFR-mutated adenocarcinomas refractory to EGFR-TKIs (Thunnissen, 2014).
 
NTRK
NTRK gene fusions encode tropomyosin receptor kinase fusion proteins that act as oncogenic drivers for solid tumors including lung, salivary gland, thyroid, and sarcoma. It is estimated that NTRK gene fusions occur in 0.2% of patients with NSCLC and do not typically overlap with other oncogenic drivers (NCCN, 2022).
 
PD-1/PD-L1
Programmed cell ligand-1 (PD-L1) is a transmembrane protein expressed on the surface of multiple tissue types, including many tumor cells. Blocking the PD-L1 protein may prevent cancer cells from inactivating T cells.
 
Tumor Mutational Burden
Tumor mutational burden, a measure of gene mutations within cancer cells, is an emerging biomarker of outcomes with immunotherapy in multiple tumor types, including lung cancer (Hellmann, 2018).
  
Targeted Treatment and Immunotherapy
FDA-approved targeted treatments and immunotherapies for the variants described above are summarized below. (Note this information is current as of October 17, 2022. FDA maintains a list of oncology drug approval notifications at https://www.fda.gov/drugs/resources-information-approved-drugs/oncology-cancer-hematologic-malignancies-approval-notifications.)
 
EGFR
    • Gefitinib (Iressa),
    • Erlotinib (Tarceva)
    • alone or in combination with ramucirumab (Cyramza)
    • Afatinib (Gilotrif)
    • Osimertinib (Tagrisso)
    • Dacomitinib (Vizimpro)
    • Amivantamab-vmjw (Rybrenant)
    • Mobocertinib (Exkivity)
ALK
    • Crizotinib (Xalkori)
    • Ceritinib (Zykadia)
    • Alectinib (Alecensa)
    • Brigatinib (Alunbrig)
    • Lorlatinib (Lorbrena)
BRAF
    • Dabrafenib (Tafinlar) alone or in combination with trametinib (Mekinist)
ROS1
    • Crizotinib (Xalkori)
    • Entrectinib (Rozlytrek)
KRAS
    • Sotorasib (Lumakras)
HER2 (ERBB2)
    • Fam-trastuzumab deruxtecan-nxki (Enhertu)
RET
    • Selpercatinib (Retevmo)
    • Pralsetinib (Gavreto)
MET
    • Capmatinib (Tabrecta)
    • Tepotinib (Tepmetko)
NTRK1
    • Larotrectinib (Vitrakvi)
    • Entrectinib (Rozlytrek)
PD-L1
    • Pembrolizumab (Keytruda)
    • Nivolumab (Opdivo) in combination with ipilimumab (Yervoy)
    • Atezolizumab (Tecentriq)
    • Cemiplimab-rwlc (Libtayo)
 
Regulatory Status
Below is a summary of the FDA-approved targeted treatments for patients with NSCLC along with the concurrently approved companion diagnostic tests. (Note this information is current as of October 17, 2022. FDA maintains a list of cleared or approved companion diagnostics at https://www.fda.gov/medical-devices/in-vitro-diagnostics/list-cleared-or-approved-companion-diagnostic-devices-in-vitro-and-imaging-tools.)
 
Treatment: Afatinib(Gilotrif)
    • Indication
      • 2013: First line for patients with metastatic NSCLC whose tumors have EGFR exon 19 deletions or exon 21 (L858R) substitutions
      • 2016: Second line for patients with metastatic squamous NSCLC
      • 2018: First line for patients with nonresistant EGFR variants other than exon 19 or exon 21 NSCLC
    • FDA-Approved Companion Diagnostic Tests
      • 2013: therascreen® EGFR Rotor-Gene Q polymerase chain reaction (RGQ PCR) kit (Qiagen)
      • 2017: FoundationOne CDx™ (Foundation Medicine)
      • 2021: ONCO/Reveal Dx Lung & Colon Cancer Assay (O/RDx-LCCA)
Treatment: Alectinib (Alecensa)
    • Indication
      • 2015: Second line for patients with ALK-positive metastatic NSCLC who have progressed on or are intolerant of crizotinib
      • 2017: Patients with ALK-positive metastatic NSCLC as detected by an FDA-approved test
    • FDA-Approved Companion Diagnostic Tests
      • 2017: FoundationOne CDx™ (Foundation Medicine)
      • 2017: Ventana ALK (D5F3) CDx Assay
      • 2020: FoundationOne Liquid CDx
Treatment: Amivantamab-vmjw (Rybrenant)
    • Indication
      • 2021: adult patients with locally advanced or metastatic NSCLC with EGFR exon 20 insertion mutations, as detected by an FDA-approved test, whose disease has progressed on or after platinum-based chemotherapy
    • FDA-Approved Companion Diagnostic Tests
      • 2021: Guardant360 CDx (0242U)
      • 2021: Oncomine™ DxTarget Test
Treatment: Atezolizumab (Tecentriq)
    • Indication
      • 2020: First-line treatment of adult patients with metastatic NSCLC whose tumors have high PD-L1 expression (PD-L1 stained 50% of tumor cells [TC 50%] or PD-L1 stained tumor-infiltrating immune cells covering 10% of the tumor area [IC 10%]), as determined by an FDA approved test, with no EGFR or ALK genomic tumor aberrations.
        • in combination with bevacizumab, paclitaxel, and carboplatin, for the first line treatment of adult patients with metastatic non-squamous NSCLC with no EGFR or ALK genomic tumor aberrations
        • in combination with paclitaxel protein-bound and carboplatin for the first line treatment of adult patients with metastatic non-squamous NSCLC with no EGFR or ALK genomic tumor aberrations
        • for the treatment of adult patients with metastatic NSCLC who have disease progression during or following platinum-containing chemotherapy.
    • FDA-Approved Companion Diagnostic Tests
      • 2020: Ventana PD-L1
Treatment: Brigatinib (Alunbrig)
    • Indication
      • 2020: Treatment of adult patients with ALK-positive metastatic NSCLC as detected by an FDA-approved test
    • FDA-Approved Companion Diagnostic Tests
      • 2020: Vysis ALK Break Apart FISH Probe Kit
Treatment: Capmatinib (Tabrecta)
    • Indication
      • 2020: Metastatic NSCLC whose tumors have a mutation that leads to MET exon 14 skipping as detected by an FDA-approved test.
    • FDA-Approved Companion Diagnostic Tests
      • 2020: FoundationOne CDx™
      • 2021: FoundationOne Liquid CDx™
Treatment: Cemiplimab-rwlc (Libtayo)
    • Indication
      • 2022: First-line treatment of patients with advanced NSCLC (locally advanced who are not candidates for surgical resection or definitive chemoradiation or metastatic) whose tumors have high PD-L1 expression (Tumor Proportion Score [TPS] > 50%) as determined by an FDA-approved test, with no EGFR, ALK or ROS1 aberrations
    • FDA-Approved Companion Diagnostic Tests
      • 2021: PD-L1 IHC 22C3 pharmDx (Dako North America, Inc.)
Treatment: Ceritinib (Zykadia)
    • Indication
      • 2014: Second line for patients with ALK-positive metastatic NSCLC who have progressed on or are intolerant of crizotinib
      • 2017: First line for patients with ALK-positive metastatic NSCLC
    • FDA-Approved Companion Diagnostic Tests
      • 2017: FoundationOne CDx™ (Foundation Medicine)
      • 2017: VENTANA ALK (D5F3) CDx Assay
Treatment: Crizotinib (Xalkori)
    • Indication
      • 2011: First line for patients with ALK-positive metastatic NSCLC
    • FDA-Approved Companion Diagnostic Tests
      • 2011: Vysis ALK Break Apart FISH Probe Kit (Abbott Laboratories)
      • 2015: Ventana ALK (D5F3) CDx Assay (Ventana Medical Systems)
      • 2017: FoundationOne CDx™ (Foundation Medicine)
      • 2017: Oncomine™ Dx Target Test (Thermo Fisher Scientific)
Treatment: Crizotinib (Xalkori)
    • Indication
      • 2016: Patients with ROS1-positive metastatic NSCLC
    • FDA-Approved Companion Diagnostic Tests
      • 2017: Oncomine™ Dx Target Test (Thermo Fisher Scientific)
Treatment: Dacomitinib (Vizimpro)
    • Indication
      • 2018: First line for patients with metastatic NSCLC with EGFR exon 19 deletion or exon 21 (L858R) substitutions
    • FDA-Approved Companion Diagnostic Tests
      • 2018: therascreen EGFR RGQ PCR Kit
      • 2021: ONCO/Reveal Dx Lung & Colon Cancer Assay (O/RDx-LCCA)
Treatment: Dabrafenib (Tafinlar) plus trametinib (Mekinist)
    • Indication
      • 2017: Used in combination for treatment of patients with metastatic NSCLC with BRAF V600E variant
    • FDA-Approved Companion Diagnostic Tests
      • 2017: Oncomine™ Dx Target Test
      • 2017: FoundationOne CDx™ (Foundation Medicine)
Treatment: Entrectinib (Rozlytrek)
    • Indication
      • 2019:
        • Adult patients with metastatic NSCLC whose tumors are ROS1-positive
        • Adult and pediatric patients 12 years of age and older with
          • solid tumors that have a NTRK gene fusion without a known acquired resistance mutation,
          • are metastatic or where surgical resection is likely to result in severe morbidity, and have progressed following treatment or have no satisfactory alternative therapy
    • FDA-Approved Companion Diagnostic Tests
      • 2022: FoundationOne CDx™ (Foundation Medicine)
Treatment: Erlotinib (Tarceva)
    • Indication
      • 2020: First-line treatment in combination with ramucirumab (Cyramza) for patients with metastatic NSCLC whose tumors have EGFR exon 19deletions or exon 21 (L858R) substitutions
      • 2013: First line for patients with metastatic NSCLC whose tumors have EGFR exon 19 deletions or exon 21 (L858R) substitutions
      • 2010: Maintenance for patients with locally advanced or metastatic NSCLC whose disease has not progressed after 4 cycles of platinum-based chemotherapy
      • 2004: Second line for patients with locally advanced or metastatic NSCLC
    • FDA-Approved Companion Diagnostic Tests
      • 2013: cobas® EGFR Mutation Test (tissue test) (Roche Diagnostics)
      • 2016: cobas® EGFR Mutation Test v2 (tissue or blood test) (Roche Diagnostics)
      • 2017: FoundationOne CDx™ (Foundation Medicine)
      • 2020: FoundationOne® Liquid CDx
      • 2021: ONCO/Reveal Dx Lung & Colon Cancer Assay (O/RDx-LCCA)
Treatment: Gefitinib (Iressa)
    • Indication
      • 2015: First line for patients with metastatic NSCLC whose tumors have EGFR exon 19 deletions or exon 21 (L858R) substitutions
      • 2003: Second line for patients with locally advanced or metastatic NSCLC
    • FDA-Approved Companion Diagnostic Tests
      • 2015: therascreen® EGFR Rotor-Gene Q polymerase chain reaction (RGQ PCR) kit
      • 2017: Oncomine™ Dx Target Test
      • 2017: FoundationOne CDx™ (Foundation Medicine)
      • 2017: cobas® EGFR Mutation Test (tissue test) (Roche Diagnostics)
      • 2020: cobas® EGFR Mutation Test v2 (tissue or plasma) (Roche Diagnostics)
      • 2020: FoundationOne® Liquid CDx
      • 2021: ONCO/Reveal Dx Lung & Colon Cancer Assay (O/RDx-LCCA)
Treatment: Larotrectinib (Vitrakvi)
    • Indication
      • 2018: Adult and pediatric patients with solid tumors that
        • have a NTRK gene fusion without a known acquired resistance mutation,
        • are metastatic or where surgical resection is likely to result in severe morbidity, and
        • have no satisfactory alternative treatments or that have progressed following treatment
    • FDA-Approved Companion Diagnostic Tests
      • 2020: FoundationOne CDx® (solid tumors, NTRK1/2/3 fusions)
Treatment: Lorlatinib (Lorbrena)
    • Indication
      • 2018: Patients with ALK-positive metastatic NSCLC whose disease has progressed on:
        • crizotinib and at least 1 other ALK inhibitor for metastatic disease; or
        • alectinib as the first ALK inhibitor therapy for metastatic disease; or
        • ceritinib as the first ALK inhibitor therapy for metastatic disease
    • FDA-Approved Companion Diagnostic Tests
      • 2021: Ventana ALK(D5F3) CDx Assay
Treatment: Mobocertinib (Exkivity)
    • Indication
      • 2021: Adult patients with locally advanced or metastatic NSCLC with EGFR exon 20 insertion mutations, as detected by an FDA-approved test, whose disease has progressed on or after platinum-based chemotherapy
    • FDA-Approved Companion Diagnostic Tests
      • 2021: Oncomine Dx Target Test
Treatment: Nivolumab (Opdivo) in combination with Ipilimumab (Yervoy)
    • Indication
      • 2020:
        • adult patients with metastatic NSCLC expressing PD-L1 (1%) as determined by an FDA-approved test, with no EGFR or ALK genomic tumor aberrations, as first-line treatment in combination with ipilimumab
        • adult patients with metastatic or recurrent NSCLC with no EGFR orALK genomic tumor aberrations as first-line treatment, in combination with ipilimumab and 2 cycles of platinum-doublet chemotherapy
        • patients with metastatic NSCLC and progression on or after platinum-based chemotherapy. Patients with EGFR or ALK genomic tumor aberrations should have disease progression on FDA-approved therapy for these aberrations prior to receiving OPDIVO.
    • FDA-Approved Companion Diagnostic Tests
      • 2020: PD-L1 IHC 28-8PharmDx
Treatment: Osimertinib (Tagrisso)
    • Indication
      • 2015: Second line for patients with metastatic NSCLC whose tumors have EGFR T790M variants as detected by an FDA-approved test, who have not responded to EGFR-blocking therapy
      • 2018: First line for patients with metastatic NSCLC whose tumors have EGFR exon 19 deletions or exon 21 L858R variants
      • 2019: EGFR exon 19 deletion and EGFR exon 21 L858R alterations
      • 2020: adjuvant therapy after tumor resection in adult patients with NSCLC whose tumors have EGFR exon 19 deletions or exon 21 L858R mutations, as detected by an FDA-approved test
    • FDA-Approved Companion Diagnostic Tests
      • 2015-2020: cobas® EGFR Mutation Test v2(tissue or plasma)
      • 2017-2019:FoundationOne CDx™ (Foundation Medicine)
      • 2020: Guardant360 CDx (0242U)
      • 2020: FoundationOne® Liquid CDx
Treatment: Pembrolizumab (Keytruda)
    • Indication
      • 2018: Monotherapy for the treatment of patients with metastatic NSCLC whose tumors express PD-L1 (TPS 1%) as determined by an FDA-approved test, with disease progression on or after platinum-containing chemotherapy; patients with EGFR or ALK genomic tumor aberrations should have disease progression on FDA-approved therapy for these aberrations prior to receiving KEYTRUDA
      • 2020: For the treatment of adult and pediatric patients with unresectable or metastatic tumor mutational burden-high (TMB-H) [10mutations/mega base (mut/Mb)] solid tumors, as determined by an FDA-approved test, that have progressed following prior treatment and who have no satisfactory alternative treatment options
    • FDA-Approved Companion Diagnostic Tests
      • 2018: PD-L1 IHC 22C3pharmDx
      • 2020: FoundationOne CDx (TMB)
Treatment: Pralsetinib (Gavreto)
    • Indication
      • 2020: Adult patients with metastatic RET fusion-positive NSCLC as detected by an FDA approved test
    • FDA-Approved Companion Diagnostic Tests
      • 2020: Oncomine Dx Target Test
Treatment: Selpercatinib (Retevmo)
    • Indication
      • 2020: Adult patients with metastatic RET fusion-positive NSCLC
    • FDA-Approved Companion Diagnostic Tests
      • 2022: Oncomine Dx Target Test
Treatment: Sotorasib (Lumakras)
    • Indication
      • 2021: Adult patients with KRAS G12C-mutated locally advanced or metastatic NSCLC, as determined by an FDA-approved test, who have received at least 1 prior systemic therapy
    • FDA-Approved Companion Diagnostic Tests
      • 2021: Therascreen KRASRGQ PCR kit
      • 2021: Guardant360 CDx (0242U)
Treatment: Tepotinib (Tepmetko)
    • Indication
      • 2021: Adult patients with metastatic NSCLC harboring MET exon 14skipping alterations.
    • FDA-Approved Companion Diagnostic Tests
      • No approved companion diagnostic
Treatment: Fam-trastuzumabderuxtecan-nxki (Enhertu)
    • Indication
      • 2022: Adult patients with unresectable or metastatic NSCLC whose tumors have activating HER2 (ERBB2) mutations, as detected by an FDA-approved test, and who have received a prior systemic therapy
    • FDA-Approved Companion Diagnostic Tests
      • 2022: Oncomine Dx Target Test
      • 2022: Guardant360 CDx (0242U)
 
Coding Guidelines:
Effective in 2013, there is a specific CPT code for testing for common variants of EGFR:
 
81235: EGFR (epidermal growth factor receptor) (e.g., non-small-cell lung cancer) gene analysis, common variants (e.g., exon 19 LREA deletion L858R, T790M, G719A, G719S, L861Q)
 
Prior to the creation of code 81235, no specific CPT codes were available, and this laboratory test would likely have been coded using a series of nonspecific genetic testing codes. One laboratory website listed the following group of CPT codes for this testing: 83907, 83900(x2), 83901(x18), 83891, 83896(x29), 83898(x6), 88381, 83914(x29), 83912-26.
 
If testing is done by immunohistochemical assay, CPT code 88342 would likely be reported. If testing is done by fluorescence in situ hybridization (FISH), CPT code 88365 would likely be reported.
 
Testing for mutations in the other genes listed above would be reported with the unlisted molecular pathology code 81479 unless a more specific code exists such as 81275 for KRAS, 81404/81405 for RET, or 81406 for BRAF.
  
Testing for NSCLC to predict sensitivity to Erlotinib or KRAS mutation to predict sensitivity to Erlotinib and Cetuximab was previously included in Archived Policies 2011004 and 2010040.

Policy/
Coverage:
In general, genetic cancer susceptibility panels are not covered, however, when coverage criteria of this or another specific policy are met, limited genetic cancer susceptibility panels are covered, including only the gene variants for which a given member qualifies, as outlined in the policy.
 
Effective October 2023
 
In general, genetic cancer susceptibility panels are not covered, however, when coverage criteria of this or another specific policy are met, limited genetic cancer susceptibility panels are covered, including only the gene variants for which a given member qualifies, as outlined in the policy.
 
Meets Primary Coverage Criteria Or Is Covered For Contracts Without Primary Coverage Criteria
 
EGFR TESTING: Analysis of tumor for somatic variants in exons 18 through 21 (eg, G719X, L858R, T790M, S6781, L861Q) within the epidermal growth factor receptor (EGFR) meets member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness to predict treatment response to an FDA-approved (e.g., erlotinib [Tarceva] alone or in combination with ramucirumab [Cyramza], gefitinib [Iressa], afatinib [Gilotrif], dacomitinib ]Vizimpro], osimertinib [Tagrisso]), or mobocertinib [Exkivity] in individuals with advanced lung adenocarcinoma, large cell carcinoma, advanced squamous cell non-small-cell lung cancer (NSCLC), and NSCLC not otherwise specified, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label.
 
Analysis of tumor tissue for somatic variants in exon 20 (eg, insertion mutations) meets member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness to predict treatment response to an FDA-approved therapy (eg, mobocertinib [Exkivity] or amivantamab [Rybrevant]) in individuals with NSCLC, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label.
 
At diagnosis, only analysis of plasma for somatic variants in exons 19 through 21 (eg, exon 19 deletions, L858R, T790M ) within the EGFR gene, using the cobas EGFR Mutation Test v2, Guardant360 CDx test (0242U), FoundationOne Liquid CDx, OncoBEAM test or InVisionFirst-Lung test to detect circulating tumor DNA (ctDNA), meets member benefit certificate primary coverage criteria as an alternative to tissue biopsy to predict treatment response to an FDA-approved therapy in individuals with advanced lung adenocarcinoma, large cell carcinoma, advanced squamous cell NSCLC, and NSCLC not otherwise specified, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label.
 
At progression, analysis of the EGFR T790M resistance variant for targeted therapy with osimertinib using ctDNA using the cobas EGFR Mutation Test v2, Guardant360 CDx test (0242U), OncoBEAM test or InVisionFirst-Lung test to detect circulating tumor DNA (ctDNA), meets member benefit certificate primary coverage criteria  in individuals with advanced lung adenocarcinoma, large cell carcinoma, advanced squamous cell NSCLC, and NSCLC, not otherwise specified when tissue biopsy to obtain new tissue is not feasible, (e.g., in those who do not have enough tissue for standard molecular testing using formalin-fixed paraffin-embedded tissue, do not have a biopsy-amenable lesion, or cannot undergo biopsy) and when the individual does not have any FDA-labeled contraindications to Osimertinib and it is intended to be used consistently with the FDA-approved label.
 
Analysis of plasma for somatic variants in exon 20 (eg, insertion mutations) within the EGFR gene using an FDA-approved companion diagnostic plasma test to detect ctDNA meets member benefit certificate primary coverage criteria as an alternative to tissue biopsy to predict treatment response to an FDA-approved therapy in individuals with  NSCLC (eg, amivantamab [Rybrevant]), if the individual does not have any FDA-labeled contraindications to the requested agent and both the agent and ctDNA test are intended to be used consistently with their FDA-approved labels
 
ALK TESTING: Analysis of somatic rearrangement variants of the anaplastic lymphoma kinase (ALK) gene meets member benefit certificate primary coverage criteria to predict treatment response to an FDA-approved ALK inhibitor therapy (e.g., crizotinib [Xalkori], ceritinib [Zykadia], alectinib [Alecensa], brigatinib [Alunbrig], or lorlatinib [Lorbrena]) in individuals with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label.
 
Analysis of plasma for somatic rearrangement variants of the ALK gene using an FDA-approved companion diagnostic plasma test to detect ctDNA meets member benefit certificate primary coverage criteria as an alternative to tissue biopsy to predict treatment response to an FDA-approved ALK inhibitor therapy in individuals with NSCLC (eg, alectinib [Alcensa]), if the individual does not have any FDA-labeled contraindications to the requested agent and both the agent and ctDNA test are intended to be used consistently with their FDA-approved labels
 
BRAF V600E TESTING: Analysis of BRAF V600E variant meets member benefit certificate primary coverage criteria to predict treatment response to an FDA-approved BRAF and/or MEK-inhibitor therapy (eg, dabrafenib [Tafinlar] and trametinib [Mekinist]), in individuals with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label.
 
ROS1 TESTING: Analysis of tumor tissue for somatic rearrangement variants of the ROS1 gene meets member benefit certificate primary coverage criteria to predict treatment response to an FDA-approved ROS1 inhibitor therapy (e.g., crizotinib [Xalkori] or entrectinib [Rozlytrek]) in individuals with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label.
 
KRAS Testing:  Analysis of tumor tissue for somatic variants of the KRAS gene meets member benefit certificate primary coverage criteria  to predict treatment response to sotorasib (Lumakras) in individuals with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label.
 
Analysis of plasma for somatic variants of the KRAS (e.g., G12C) using an FDA- approved companion diagnostic plasma test to detect ctDNA meets member benefit certificate primary coverage to predict treatment response to sotorasib (Lumakras) in individuals with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded if the individual does not have any FDA-labeled contraindications to the requested agent and both the agent and ctDNA test are intended to be used consistently with their FDA-approved labels.
  
HER2 Testing: Analysis of tumor tissue for somatic alterations in the HER2(ERBB2) gene meets member benefit certificate primary coverage criteria to predict treatment response to an FDA-approved therapy (eg, fam-trastuzumab deruxtecan-nxki [Enhertu]) in individuals with unresectable or metastatic NSCLC, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label.
 
Analysis of plasma for somatic alterations in the HER2(ERBB2) gene using an FDA-approved companion diagnostic plasma test to detect ctDNA meets member benefit certificate primary coverage criteria as an alternative to tissue biopsy to predict treatment response to an FDA-approved therapy (eg, fam-trastuzumab deruxtecan-nxki [Enhertu]) in individuals with unresectable or metastatic NSCLC, if the individual does not have any FDA-labeled contraindications to the requested agent and both the agent and ctDNA test are intended to be used consistently with their FDA-approved labels.
 
NTRK GENE FUSION TESTING:  Analysis of tumor tissue for somatic by immunohistochemistry (IHC) of NTRK gene fusions meets member benefit certificate primary coverage criteria to predict treatment response to entrectinib (Rozlytrek) or Larotrectinib (Vitrakvi) in individuals with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label.
 
RET REARRANGEMENT TESTING: Analysis of tumor tissue for somatic alterations in the RET gene meets member benefit certificate primary coverage criteria to predict treatment response to pralsetinib (Gavreto) or selpercatinib (Retevmo) in individuals with metastatic NSCLC if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label.
 
MET EXON 14 SKIPPING ALTERATION: Analysis of tumor tissue for somatic alteration that leads to MET exon 14 skipping meets member benefit certificate primary coverage criteria to predict treatment response to capmatinib (Tabrecta) in individuals with metastatic NSCLC, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label to predict treatment response to capmatinib (Tabrecta). in individuals with metastatic NSCLC.
 
Analysis of plasma for somatic alteration that leads to MET exon 14 skipping using an FDA-approved companion diagnostic plasma test specimens to detect ctDNA meets member benefit certificate primary coverage criteria as an alternative to tissue biopsy to predict treatment response to MET inhibitor therapy (e.g., capmatinib [Tabrecta]) in patients with NSCLC, if the individual does not have any FDA-labeled contraindications to the requested agent and both the agent and ctDNA test are intended to be used consistently with their FDA-approved labels.
 
PD-L1 TESTING: PD-L1 testing of tumor tissue meets member benefit certificate primary coverage criteria to predict treatment response to an FDA-approved therapy (e.g., atezolizumab [Tecentriq], nivolumab [Opdivo] in combination with ipilimumab [Yervoy], pembrolizumab [Keytruda],  or cemiplimab-rwlc [Libtayo]) in individuals with NSCLC, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label
 
Plasma Testing When Tissue is Insufficient: Plasma tests for oncogenic driver variants meeting primary coverage criteria on tissue biopsy when there is insufficient tissue meets member benefit certificate primary coverage criteria  to predict treatment response to targeted therapy for individuals meeting all the following criteria:
 
        • Individual does not have sufficient tissue for standard molecular testing using formalin-fixed paraffin-embedded tissue; AND
        • Follow-up tissue-based analysis is planned should no driver variant be identified via plasma testing.
 
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
 
Testing for the following does not meet member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness in improving health outcomes in any clinical situations or other applications not addressed above or in other policies for individuals with NSCLC:
 
    • Analysis of somatic variants in the EGFR gene in tissue or plasma, including variants within exons 22 to 24;
    • Analysis of somatic rearrangement variants of the ALK gene in tissue or plasma;
    • Analysis of tumor tissue for the somatic BRAF V600E variant in all other situations;
    • Analysis of plasma for the somatic BRAF V600E variant to detect ctDNA as an alternative to tissue biopsy to predict treatment response to BRAF and/or MEK inhibitor therapy (eg, dabrafenib [Tafinlar], trametinib [Mekinist]) in individuals with NSCLC;
    • Analysis of tumor tissue for somatic rearrangement variants of the ROS1 gene in all other situations;
    • Analysis of plasma for somatic rearrangement variants of the ROS1 gene to detect ctDNA as an alternative to tissue biopsy to predict treatment response to ROS1 inhibitor therapy (e.g., crizotinib [Xalkori] or entrectinib [Rozlytrek])) in individuals with NSCLC;
    • All other uses of analysis of somatic variants of the KRAS gene in tissue or plasma;
    • All other uses of analysis of somatic variants of the HER2 (ERBB2) gene in tissue or plasma;
    • Analysis of plasma for somatic NTRK gene fusions and analysis of somatic alterations in the NTRK gene fusions in all other situations;
    • Analysis of tumor tissue for somatic alterations in the RET gene in all other situations;
    • Analysis of plasma for somatic alterations of the RET gene to detect ctDNA as an alternative to tissue biopsy to predict treatment response to RET inhibitor therapy (eg, selpercatinib [Retevmo], pralsetinib [Gavreto]) in individuals with NSCLC;
    • Analysis of somatic variants of the MET gene in tissue or plasma in all other situations;
    • PD-L1 testing in all other situations;
    • Analysis of tumor mutational burden to predict response to immunotherapy in individuals with NSCLC;
    • Plasma tests for oncogenic driver variants when there is insufficient tissue to predict treatment response to targeted therapy for individuals not meeting the above criteria.
 
For members with contracts without primary coverage criteria, the use of the above tests for any clinical situation or application not addressed in this or other policies for individuals with NSCLC are considered investigational. Investigational services are specific contract exclusions in most member benefit certificates of coverage.
 
Augmentative algorithmic analysis of digitized whole slide imaging (e.g., LungOI, 0414U) does not meet member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness in improving health outcomes for individuals with NSCLC.
 
For members with contracts without primary coverage criteria, augmentative algorithmic analysis of digitized whole slide imaging (e.g., LungOI, 0414U) for individuals with NSCLC is considered investigational. Investigational services are specific contract exclusions in most member benefit certificates of coverage.
 
Effective June 2023 through September 2023
 
In general, genetic cancer susceptibility panels are not covered, however, when coverage criteria of this or another specific policy are met, limited genetic cancer susceptibility panels are covered, including only the gene variants for which a given member qualifies, as outlined in the policy.
 
Meets Primary Coverage Criteria Or Is Covered For Contracts Without Primary Coverage Criteria
 
EGFR TESTING: Analysis of tumor for somatic variants in exons 18 through 21 (eg, G719X, L858R, T790M, S6781, L861Q) within the epidermal growth factor receptor (EGFR) meets member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness to predict treatment response to an FDA-approved (e.g., erlotinib [Tarceva] alone or in combination with ramucirumab [Cyramza], gefitinib [Iressa], afatinib [Gilotrif], dacomitinib ]Vizimpro], osimertinib [Tagrisso]), or mobocertinib [Exkivity] in individuals with advanced lung adenocarcinoma, large cell carcinoma, advanced squamous cell non-small-cell lung cancer (NSCLC), and NSCLC not otherwise specified, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label.
 
Analysis of tumor tissue for somatic variants in exon 20 (eg, insertion mutations) meets member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness to predict treatment response to an FDA-approved therapy (eg, mobocertinib [Exkivity] or amivantamab [Rybrevant]) in individuals with NSCLC, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label
 
At diagnosis, only analysis of plasma for somatic variants in exons 19 through 21 (eg, exon 19 deletions, L858R, T790M ) within the EGFR gene, using the cobas EGFR Mutation Test v2, Guardant360 CDx test, FoundationOne Liquid CDx, OncoBEAM test or InVisionFirst-Lung test to detect circulating tumor DNA (ctDNA), meets member benefit certificate primary coverage criteria as an alternative to tissue biopsy to predict treatment response to an FDA-approved therapy in individuals with advanced lung adenocarcinoma, large cell carcinoma, advanced squamous cell NSCLC, and NSCLC not otherwise specified, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label.
 
At progression, analysis of the EGFR T790M resistance variant for targeted therapy with osimertinib using ctDNA using the cobas EGFR Mutation Test v2, Guardant360 CDx test, OncoBEAM test or InVisionFirst-Lung test to detect circulating tumor DNA (ctDNA), meets member benefit certificate primary coverage criteria  in individuals with advanced lung adenocarcinoma, large cell carcinoma, advanced squamous cell NSCLC, and NSCLC, not otherwise specified when tissue biopsy to obtain new tissue is not feasible, (e.g., in those who do not have enough tissue for standard molecular testing using formalin-fixed paraffin-embedded tissue, do not have a biopsy-amenable lesion, or cannot undergo biopsy) and when the individual does not have any FDA-labeled contraindications to Osimertinib and it is intended to be used consistently with the FDA-approved label.
 
Analysis of plasma for somatic variants in exon 20 (eg, insertion mutations) within the EGFR gene using an FDA-approved companion diagnostic plasma test to detect ctDNA meets member benefit certificate primary coverage criteria as an alternative to tissue biopsy to predict treatment response to an FDA-approved therapy in individuals with  NSCLC (eg, amivantamab [Rybrevant]), if the individual does not have any FDA-labeled contraindications to the requested agent and both the agent and ctDNA test are intended to be used consistently with their FDA-approved labels
 
ALK TESTING: Analysis of somatic rearrangement variants of the anaplastic lymphoma kinase (ALK) gene meets member benefit certificate primary coverage criteria to predict treatment response to an FDA-approved ALK inhibitor therapy (e.g., crizotinib [Xalkori], ceritinib [Zykadia], alectinib [Alecensa], brigatinib [Alunbrig], or lorlatinib [Lorbrena]) in individuals with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label.
 
Analysis of plasma for somatic rearrangement variants of the ALK gene using an FDA-approved companion diagnostic plasma test to detect ctDNA meets member benefit certificate primary coverage criteria as an alternative to tissue biopsy to predict treatment response to an FDA-approved ALK inhibitor therapy in individuals with NSCLC (eg, alectinib [Alcensa]), if the individual does not have any FDA-labeled contraindications to the requested agent and both the agent and ctDNA test are intended to be used consistently with their FDA-approved labels
 
BRAF V600E TESTING: Analysis of BRAF V600E variant meets member benefit certificate primary coverage criteria to predict treatment response to an FDA-approved BRAF and/or MEK-inhibitor therapy (eg, dabrafenib [Tafinlar] and trametinib [Mekinist]), in individuals with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label.
 
ROS1 TESTING: Analysis of tumor tissue for somatic rearrangement variants of the ROS1 gene meets member benefit certificate primary coverage criteria to predict treatment response to an FDA-approved ROS1 inhibitor therapy (e.g., crizotinib [Xalkori] or entrectinib [Rozlytrek]) in individuals with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label.
 
KRAS Testing:  Analysis of tumor tissue for somatic variants of the KRAS gene meets member benefit certificate primary coverage criteria  to predict treatment response to sotorasib (Lumakras) in individuals with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label.
 
Analysis of plasma for somatic variants of the KRAS (e.g., G12C) using an FDA- approved companion diagnostic plasma test to detect ctDNA meets member benefit certificate primary coverage to predict treatment response to sotorasib (Lumakras) in individuals with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded if the individual does not have any FDA-labeled contraindications to the requested agent and both the agent and ctDNA test are intended to be used consistently with their FDA-approved labels.
  
HER2 Testing: Analysis of tumor tissue for somatic alterations in the HER2(ERBB2) gene meets member benefit certificate primary coverage criteria to predict treatment response to an FDA-approved therapy (eg, fam-trastuzumab deruxtecan-nxki [Enhertu]) in individuals with unresectable or metastatic NSCLC, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label.
 
Analysis of plasma for somatic alterations in the HER2(ERBB2) gene using an FDA-approved companion diagnostic plasma test to detect ctDNA meets member benefit certificate primary coverage criteria as an alternative to tissue biopsy to predict treatment response to an FDA-approved therapy (eg, fam-trastuzumab deruxtecan-nxki [Enhertu]) in individuals with unresectable or metastatic NSCLC, if the individual does not have any FDA-labeled contraindications to the requested agent and both the agent and ctDNA test are intended to be used consistently with their FDA-approved labels.
 
NTRK GENE FUSION TESTING:  Analysis of tumor tissue for somatic by immunohistochemistry (IHC) of NTRK gene fusions meets member benefit certificate primary coverage criteria to predict treatment response to entrectinib (Rozlytrek) or Larotrectinib (Vitrakvi) in individuals with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label.
 
RET REARRANGEMENT TESTING: Analysis of tumor tissue for somatic alterations in the RET gene meets member benefit certificate primary coverage criteria to predict treatment response to pralsetinib (Gavreto) or selpercatinib (Retevmo) in individuals with metastatic NSCLC if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label.
 
MET EXON 14 SKIPPING ALTERATION: Analysis of tumor tissue for somatic alteration that leads to MET exon 14 skipping meets member benefit certificate primary coverage criteria to predict treatment response to capmatinib (Tabrecta) in individuals with metastatic NSCLC, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label to predict treatment response to capmatinib (Tabrecta). in individuals with metastatic NSCLC.
 
Analysis of plasma for somatic alteration that leads to MET exon 14 skipping using an FDA-approved companion diagnostic plasma test specimens to detect ctDNA meets member benefit certificate primary coverage criteria as an alternative to tissue biopsy to predict treatment response to MET inhibitor therapy (e.g., capmatinib [Tabrecta]) in patients with NSCLC, if the individual does not have any FDA-labeled contraindications to the requested agent and both the agent and ctDNA test are intended to be used consistently with their FDA-approved labels.
 
PD-L1 TESTING: PD-L1 testing of tumor tissue meets member benefit certificate primary coverage criteria to predict treatment response to an FDA-approved therapy (e.g., atezolizumab [Tecentriq], nivolumab [Opdivo] in combination with ipilimumab [Yervoy], pembrolizumab [Keytruda],  or cemiplimab-rwlc [Libtayo]) in individuals with NSCLC, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label
 
Plasma Testing When Tissue is Insufficient: Plasma tests for oncogenic driver variants meeting primary coverage criteria on tissue biopsy when there is insufficient tissue meets member benefit certificate primary coverage criteria  to predict treatment response to targeted therapy for individuals meeting all the following criteria:
 
    • Individual does not have sufficient tissue for standard molecular testing using formalin-fixed paraffin-embedded tissue; AND
    • Follow-up tissue-based analysis is planned should no driver variant be identified via plasma testing.
 
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
 
Testing for the following does not meet member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness in improving health outcomes in any clinical situations or other applications not addressed above or in other policies for individuals with NSCLC:
 
    • Analysis of somatic variants in the EGFR gene in tissue or plasma, including variants within exons 22 to 24;
    • Analysis of somatic rearrangement variants of the ALK gene in tissue or plasma;
    • Analysis of tumor tissue for the somatic BRAF V600E variant in all other situations;
    • Analysis of plasma for the somatic BRAF V600E variant to detect ctDNA as an alternative to tissue biopsy to predict treatment response to BRAF and/or MEK inhibitor therapy (eg, dabrafenib [Tafinlar], trametinib [Mekinist]) in individuals with NSCLC;
    • Analysis of tumor tissue for somatic rearrangement variants of the ROS1 gene in all other situations;
    • Analysis of plasma for somatic rearrangement variants of the ROS1 gene to detect ctDNA as an alternative to tissue biopsy to predict treatment response to ROS1 inhibitor therapy (e.g., crizotinib [Xalkori] or entrectinib [Rozlytrek])) in individuals with NSCLC;
    • All other uses of analysis of somatic variants of the KRAS gene in tissue or plasma;
    • All other uses of analysis of somatic variants of the HER2 (ERBB2) gene in tissue or plasma;
    • Analysis of plasma for somatic NTRK gene fusions and analysis of somatic alterations in the NTRK gene fusions in all other situations;
    • Analysis of tumor tissue for somatic alterations in the RET gene in all other situations;
    • Analysis of plasma for somatic alterations of the RET gene to detect ctDNA as an alternative to tissue biopsy to predict treatment response to RET inhibitor therapy (eg, selpercatinib [Retevmo], pralsetinib [Gavreto]) in individuals with NSCLC;
    • Analysis of somatic variants of the MET gene in tissue or plasma in all other situations;
    • PD-L1 testing in all other situations;
    • Analysis of tumor mutational burden to predict response to immunotherapy in individuals with NSCLC;
    • Plasma tests for oncogenic driver variants when there is insufficient tissue to predict treatment response to targeted therapy for individuals not meeting the above criteria.
 
For members with contracts without primary coverage criteria, the use of the above tests for any clinical situation or application not addressed in this or other policies for individuals with NSCLC are considered investigational. Investigational services are specific contract exclusions in most member benefit certificates of coverage.
 
Effective November 2022 through May 2023
 
Meets Primary Coverage Criteria Or Is Covered For Contracts Without Primary Coverage Criteria
 
EGFR TESTING: Analysis of somatic variants in exons 18 through 21 (eg, G719X, L858R, T790M, S6781, L861Q) within the epidermal growth factor receptor (EGFR) meets member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness to predict treatment response to an FDA-approved (e.g., erlotinib [Tarceva] alone or in combination with ramucirumab [Cyramza], gefitinib [Iressa], afatinib [Gilotrif], dacomitinib ]Vizimpro], osimertinib [Tagrisso]), or mobocertinib [Exkivity] in individuals with advanced lung adenocarcinoma, large cell carcinoma, advanced squamous cell non-small-cell lung cancer (NSCLC), and NSCLC not otherwise specified, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label.
 
Analysis of tumor tissue for somatic variants in exon 20 (eg, insertion mutations) meets member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness to predict treatment response to an FDA-approved therapy (eg, mobocertinib [Exkivity] or amivantamab [Rybrevant]) in individuals with NSCLC, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label
 
At diagnosis, only analysis of somatic variants in exons 19 through 21 (eg, exon 19 deletions, L858R, T790M ) within the EGFR gene, using the cobas EGFR Mutation Test v2, Guardant360 CDx test, FoundationOne Liquid CDx, OncoBEAM test or InVisionFirst-Lung test to detect circulating tumor DNA (ctDNA), meets member benefit certificate primary coverage criteria as an alternative to tissue biopsy to predict treatment response to an FDA-approved therapy in individuals with advanced lung adenocarcinoma, large cell carcinoma, advanced squamous cell NSCLC, and NSCLC not otherwise specified, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label.
 
At progression, analysis of the EGFR T790M resistance variant for targeted therapy with osimertinib using ctDNA using the cobas EGFR Mutation Test v2, Guardant360 CDx test, OncoBEAM test or InVisionFirst-Lung test to detect circulating tumor DNA (ctDNA), meets member benefit certificate primary coverage criteria  in individuals with advanced lung adenocarcinoma, large cell carcinoma, advanced squamous cell NSCLC, and NSCLC, not otherwise specified when tissue biopsy to obtain new tissue is not feasible, (e.g., in those who do not have enough tissue for standard molecular testing using formalin-fixed paraffin-embedded tissue, do not have a biopsy-amenable lesion, or cannot undergo biopsy) and when the individual does not have any FDA-labeled contraindications to Osimertinib and it is intended to be used consistently with the FDA-approved label.
 
Analysis of plasma for somatic variants in exon 20 (eg, insertion mutations) within the EGFR gene using an FDA-approved companion diagnostic plasma test to detect ctDNA meets member benefit certificate primary coverage criteria as an alternative to tissue biopsy to predict treatment response to an FDA-approved therapy in individuals with  NSCLC (eg, amivantamab [Rybrevant]), if the individual does not have any FDA-labeled contraindications to the requested agent and both the agent and ctDNA test are intended to be used consistently with their FDA-approved labels
 
ALK TESTING: Analysis of somatic rearrangement variants of the anaplastic lymphoma kinase (ALK) gene meets member benefit certificate primary coverage criteria to predict treatment response to an FDA-approved ALK inhibitor therapy (e.g., crizotinib [Xalkori], ceritinib [Zykadia], alectinib [Alecensa], brigatinib [Alunbrig], or lorlatinib [Lorbrena]) in individuals with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label.
 
Analysis of plasma for somatic rearrangement variants of the ALK gene using an FDA-approved companion diagnostic plasma test to detect ctDNA meets member benefit certificate primary coverage criteria as an alternative to tissue biopsy to predict treatment response to an FDA-approved ALK inhibitor therapy in individuals with NSCLC (eg, alectinib [Alcensa]), if the individual does not have any FDA-labeled contraindications to the requested agent and both the agent and ctDNA test are intended to be used consistently with their FDA-approved labels
 
BRAF V600E TESTING: Analysis of BRAF V600E variant meets member benefit certificate primary coverage criteria to predict treatment response to an FDA-approved BRAF and/or MEK-inhibitor therapy (eg, dabrafenib [Tafinlar] and trametinib [Mekinist]), in individuals with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label.
 
ROS1 TESTING: Analysis of tumor tissue for somatic rearrangement variants of the ROS1 gene meets member benefit certificate primary coverage criteria to predict treatment response to an FDA-approved ROS1 inhibitor therapy (e.g., crizotinib [Xalkori] or entrectinib [Rozlytrek]) in individuals with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label.
 
KRAS Testing:  Analysis of tumor tissue for somatic variants of the KRAS gene meets member benefit certificate primary coverage criteria  to predict treatment response to sotorasib (Lumakras) in individuals with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label.
 
Analysis of plasma for somatic variants of the KRAS (e.g., G12C) using an FDA- approved companion diagnostic plasma test to detect ctDNA meets member benefit certificate primary coverage to predict treatment response to sotorasib (Lumakras) in individuals with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded if the individual does not have any FDA-labeled contraindications to the requested agent and both the agent and ctDNA test are intended to be used consistently with their FDA-approved labels.
  
HER2 Testing: Analysis of tumor tissue for somatic alterations in the HER2(ERBB2) gene meets member benefit certificate primary coverage criteria to predict treatment response to an FDA-approved therapy (eg, fam-trastuzumab deruxtecan-nxki [Enhertu]) in individuals with unresectable or metastatic NSCLC, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label.
 
Analysis of plasma for somatic alterations in the HER2(ERBB2) gene using an FDA-approved companion diagnostic plasma test to detect ctDNA meets member benefit certificate primary coverage criteria as an alternative to tissue biopsy to predict treatment response to an FDA-approved therapy (eg, fam-trastuzumab deruxtecan-nxki [Enhertu]) in individuals with unresectable or metastatic NSCLC, if the individual does not have any FDA-labeled contraindications to the requested agent and both the agent and ctDNA test are intended to be used consistently with their FDA-approved labels.
 
NTRK GENE FUSION TESTING:  Analysis by immunohistochemistry (IHC) of NTRK gene fusions meets member benefit certificate primary coverage criteria to predict treatment response to entrectinib or larotrectinib in individuals with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded.
 
RET REARRANGEMENT TESTING: Analysis of tumor tissue for somatic alterations in the RET gene meets member benefit certificate primary coverage criteria to predict treatment response to pralsetinib (Gavreto) or selpercatinib (Retevmo) in individuals with metastatic NSCLC if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label.
 
MET EXON 14 SKIPPING ALTERATION: Analysis of tumor tissue for somatic alteration that leads to MET exon 14 skipping meets member benefit certificate primary coverage criteria to predict treatment response to capmatinib (Tabrecta) in individuals with metastatic NSCLC, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label to predict treatment response to capmatinib (Tabrecta) in individuals with metastatic NSCLC.
 
Analysis of plasma for somatic alteration that leads to MET exon 14 skipping using an FDA-approved companion diagnostic plasma test specimens to detect ctDNA meets member benefit certificate primary coverage criteria as an alternative to tissue biopsy to predict treatment response to MET inhibitor therapy (e.g., capmatinib [Tabrecta]) in patients with NSCLC, if the individual does not have any FDA-labeled contraindications to the requested agent and both the agent and ctDNA test are intended to be used consistently with their FDA-approved labels.
 
PD-L1 TESTING: PD-L1 testing of tissue meets member benefit certificate primary coverage criteria to predict treatment response to an FDA-approved therapy (e.g., atezolizumab [Tecentriq], nivolumab [Opdivo] in combination with ipilimumab [Yervoy], pembrolizumab [Keytruda],  or cemiplimab-rwlc [Libtayo]) in individuals with NSCLC, if the individual does not have any FDA-labeled contraindications to the requested agent and the agent is intended to be used consistently with the FDA-approved label
 
Plasma Testing When Tissue is Insufficient: Plasma tests for oncogenic driver variants meeting primary coverage criteria on tissue biopsy when there is insufficient tissue meets member benefit certificate primary coverage criteria  to predict treatment response to targeted therapy for individuals meeting all the following criteria:
 
· Individual does not have sufficient tissue for standard molecular testing using formalin-fixed paraffin-embedded tissue; AND
· Follow-up tissue-based analysis is planned should no driver variant be identified via plasma testing.
 
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
 
Testing for the following does not meet member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness in improving health outcomes in any clinical situations or other applications not addressed above or other policies for individuals with NSCLC:
 
· Analysis of somatic variants in the EGFR gene in tissue or plasma, including variants within exons 22 to 24;
· Analysis of somatic rearrangement variants of the ALK gene in tissue or plasma;
· Analysis of tumor tissue for the somatic BRAF V600E variant in all other situations;
· Analysis of plasma for the somatic BRAF V600E variant to detect ctDNA as an alternative to tissue biopsy to predict treatment response to BRAF and/or MEK inhibitor therapy (eg, dabrafenib [Tafinlar], trametinib [Mekinist]) in individuals with NSCLC;
· Analysis of tumor tissue for somatic rearrangement variants of the ROS1 gene in all other situations;
· Analysis of plasma for somatic rearrangement variants of the ROS1 gene to detect ctDNA as an alternative to tissue biopsy to predict treatment response to ROS1 inhibitor therapy (e.g., crizotinib [Xalkori] or entrectinib [Rozlytrek])) in individuals with NSCLC;
· All other uses of analysis of somatic variants of the KRAS gene in tissue or plasma;
· All other uses of analysis of somatic variants of the HER2 (ERBB2) gene in tissue or plasma;
· Analysis of somatic NTRK gene fusions in all other situations;
· Analysis of tumor tissue for somatic alterations in the RET gene in all other situations;
· Analysis of plasma for somatic alterations of the RET gene to detect ctDNA as an alternative to tissue biopsy to predict treatment response to RET inhibitor therapy (eg, selpercatinib [Retevmo], pralsetinib [Gavreto]) in individuals with NSCLC;
· Analysis of somatic variants of the MET gene in tissue or plasma in all other situations;
· PD-L1 testing in all other situations;
· Analysis of tumor mutational burden to predict response to immunotherapy in individuals with NSCLC;
· Plasma tests for oncogenic driver variants when there is insufficient tissue to predict treatment response to targeted therapy for individuals not meeting the above criteria.
 
For members with contracts without primary coverage criteria, the use of the above tests for any clinical situation or application not addressed in this or other policies for individuals with NSCLC are considered investigational. Investigational services are specific contract exclusions in most member benefit certificates of coverage.
 
Effective October 1, 2022 through October 31, 2022
 
In general, genetic cancer susceptibility panels are not covered, however, when coverage criteria of this or another specific policy are met, limited genetic cancer susceptibility panels are covered, including only the gene variants for which a given member qualifies, as outlined in the policy.
 
Meets Primary Coverage Criteria Or Is Covered For Contracts Without Primary Coverage Criteria
 
EGFR TESTING: Analysis of somatic variants in exons 18 through 21 (e.g., G719X, L858R, T790M, S6781, L861Q) within the epidermal growth factor receptor (EGFR) meets member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness to predict treatment response to an EGFR tyrosine kinase inhibitor therapy (e.g., erlotinib [Tarceva], gefitinib [Iressa], afatinib [Gilotrif], osimertinib [Tagrisso]), or mobocertinib [Exkivity] in individuals with advanced lung adenocarcinoma, large cell carcinoma, advanced squamous cell non-small-cell lung cancer, and non-small-cell lung cancer not otherwise specified.
 
Use of a circulating tumor DNA test (cobas® EGFR Mutation Test v2) to detect the above specific mutations of the EGFR gene meets member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness when the volume of formalin-fixed paraffin-embedded tumor tissue (FFPET) available for testing is insufficient and the other above criteria are met.
 
KRAS Testing: Analysis of the KRAS gene for the presence of KRAS G12C mutation meets member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness to predict treatment response to sotorasib (Lumakras) in individuals with locally advanced or metastatic non-small cell lung cancer (NSCLC) who have received one prior systemic treatment.
 
ALK TESTING: Analysis of somatic rearrangement mutations of the ALK gene meets member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness to predict treatment response to ALK inhibitor therapy (e.g., crizotinib, alectinib, or brigatinib in individuals with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded.
 
BRAF V600E TESTING: Analysis of BRAF V600E variant meets member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness to predict treatment response to BRAF or MEK-inhibitor therapy (e.g., dabrafenib [Tafinlar] and trametinib [Mekinist]) in individuals with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded, and to predict treatment response to BRAF and MEK-inhibitor combination therapy in individuals with BRAF V600E–mutated non–small cell lung cancer.
 
ROS1 TESTING: Analysis of somatic rearrangement variants of the ROS1 gene meets member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness to predict treatment response to ALK inhibitor therapy (crizotinib [Xalkori]) in individuals with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded.
 
NTRK GENE FUSION TESTING: Analysis by immunohistochemistry (IHC) of NTRK gene fusions meets member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness to predict treatment response to entrectinib or larotrectinib in individuals with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded.
 
RET REARRANGEMENT TESTING: Analysis of genetic alteration in the RET gene meets member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness to predict treatment response to pralsetinib (Gavreto) or selpercatinib (Retevmo) in individuals with metastatic NSCLC.
 
MET EXON 14 SKIPPING ALTERATION: Analysis of genetic alteration that leads to MET exon 14 skipping gene meets member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness to predict treatment response to capmatinib (Tabrecta) in individuals with metastatic NSCLC.
 
PD-L1 TESTING: PD-L1 testing gene meets member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness to predict treatment response to atezolizumab (Tecentriq), nivolumab (Opdivo) in combination with ipilimumab (Yervoy) or pembrolizumab (Keytruda) in individuals with metastatic NSCLC.
 
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
 
The use of the above tests for any clinical situation or application not addressed in this or other policies for individuals with NSCLC, including but not limited to the following, does not meet member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness in improving health outcomes:
 
    • Analysis of other EGFR variants within exons 22 to 24;
    • Analysis of somatic mutations of the KRAS gene as a technique to predict treatment nonresponse to anti-EGFR therapy with the tyrosine-kinase inhibitors (e.g., erlotinib) and for the use of the anti-EGFR monoclonal antibody (e.g., cetuximab);
    • Analysis of genetic alterations in the HER2 gene for targeted therapy in individuals with NSCLC;
    • Testing for genetic alterations in the genes RET and MET for targeted therapy in individuals with NSCLC;
    • Analysis of somatic rearrangement mutations of the ALK gene.
 
For members with contracts without primary coverage criteria, the use of the above tests for any clinical situation or application not addressed in this or other policies for individuals with NSCLC, including but not limited to the following, are considered investigational. Investigational services are specific contract exclusions in most member benefit certificates of coverage.
 
    • Analysis of other EGFR variants within exons 22 to 24;
    • Analysis of somatic mutations of the KRAS gene as a technique to predict treatment nonresponse to anti-EGFR therapy with the tyrosine-kinase inhibitors (e.g., erlotinib) and for the use of the anti-EGFR monoclonal antibody (e.g., cetuximab);
    • Analysis of genetic alterations in the HER2 gene for targeted therapy in individuals with NSCLC;
    • Testing for genetic alterations in the genes RET and MET for targeted therapy in individuals with NSCLC;
    • Analysis of somatic rearrangement mutations of the ALK gene.
 
Effective November 2021 to September 2022
 
In general, genetic cancer susceptibility panels are not covered, however, when coverage criteria of this or another specific policy are met, limited genetic cancer susceptibility panels are covered, including only the gene variants for which a given member qualifies, as outlined in the policy.
 
Meets Primary Coverage Criteria Or Is Covered For Contracts Without Primary Coverage Criteria
 
EGFR TESTING: Analysis of somatic variants in exons 18 through 21 (eg, G719X, L858R, T790M, S6781, L861Q) within the epidermal growth factor receptor (EGFR) meets member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness to predict treatment response to an EGFR tyrosine kinase inhibitor therapy (eg, erlotinib [Tarceva], gefitinib [Iressa], afatinib [Gilotrif], osimertinib [Tagrisso]), or mobocertinib [Exkivity] in patients with advanced lung adenocarcinoma, large cell carcinoma, advanced squamous cell non-small-cell lung cancer, and non-small-cell lung cancer not otherwise specified.
 
Use of a circulating tumor DNA test (cobas® EGFR Mutation Test v2 ) to detect the above specific mutations of the EGFR gene meets primary coverage criteria when the volume of formalin-fixed paraffin-embedded tumor tissue (FFPET) available for testing is insufficient and the other above criteria are met.
 
KRAS Testing: Analysis of the KRAS gene for the presence of KRAS G12C mutation meets member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness in improving health outcomes to predict treatment response to sotorasib (Lumakras) in patients with locally advanced or metastatic non-small cell lung cancer (NSCLC) who have received one prior systemic treatment.
 
ALK TESTING: Analysis of somatic rearrangement mutations of the ALK gene meets member benefit certificate primary coverage criteria to predict treatment response to ALK inhibitor therapy (eg, crizotinib, alectinib, or brigatinib in patients with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded.
 
BRAF V600E TESTING: Analysis of BRAF V600E variant meets member benefit certificate primary coverage criteria to predict treatment response to BRAF or MEK-inhibitor therapy (eg, dabrafenib [Tafinlar] and trametinib [Mekinist]), in patients with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded.
 
ROS1 TESTING: Analysis of somatic rearrangement variants of the ROS1 gene meets member benefit certificate primary coverage criteria to predict treatment response to ALK inhibitor therapy (crizotinib [Xalkori]) in patients with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded.
 
NTRK GENE FUSION TESTING:  Analysis by immunohistochemistry (IHC) of NTRK gene fusions meets member benefit certificate primary coverage criteria to predict treatment response to entrectinib or larotrectinib in patients with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded.
 
RET REARRANGEMENT TESTING: Analysis of genetic alteration in the RET gene meets member benefit certificate primary coverage criteria to predict treatment response to pralsetinib (Gavreto) or selpercatinib (Retevmo) in patients with metastatic NSCLC.
 
MET EXON 14 SKIPPING ALTERATION: Analysis of genetic alteration that leads to MET exon 14 skipping gene meets member benefit certificate primary coverage criteria to predict treatment response to capmatinib (Tabrecta) in patients with metastatic NSCLC.
 
PD-L1 TESTING: PD-L1 testing gene meets member benefit certificate primary coverage criteria to predict treatment response to atezolizumab (Tecentriq), nivolumab (Opdivo) in combination with ipilimumab (Yervoy) or pembrolizumab (Keytruda) in patients with metastatic NSCLC.
 
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
 
Testing for the following indications does not meet member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness in improving health outcomes in any clinical situations or other applications not described above for NSCLC:
 
    • Analysis of other EGFR variants within exons 22 to 24;
    • Analysis of somatic mutations of the KRAS gene as a technique to predict treatment nonresponse to anti-EGFR therapy with the tyrosine-kinase inhibitors (eg, erlotinib) and for the use of the anti-EGFR monoclonal antibody (eg, cetuximab);
    • Analysis of genetic alterations in the HER2 gene for targeted therapy in patients with NSCLC;
    • Testing for genetic alterations in the genes RET and MET for targeted therapy in patients with NSCLC;
    • Analysis of somatic rearrangement mutations of the ALK gene.
 
For members with contracts without primary coverage criteria testing for the following indications are considered investigational. Investigational services are specific contract exclusions in most member benefit certificates of coverage in any clinical situations or other applications not described above for NSCLC:
 
    • Analysis of other EGFR variants within exons 22 to 24;
    • Analysis of somatic mutations of the KRAS gene as a technique to predict treatment nonresponse to anti-EGFR therapy with the tyrosine-kinase inhibitors (eg, erlotinib) and for the use of the anti-EGFR monoclonal antibody (eg, cetuximab);
    • Testing for genetic alterations in the genes RET and MET for targeted therapy in patients with NSCLC;
    • Analysis of genetic alterations in the HER2 gene for targeted therapy in patients with NSCLC;  
    • Analysis of somatic rearrangement mutations of the ALK gene.
 
Effective July 2021 to October 2021
 
In general, genetic cancer susceptibility panels are not covered, however, when coverage criteria of this or another specific policy are met, limited genetic cancer susceptibility panels are covered, including only the gene variants for which a given member qualifies, as outlined in the policy.
 
Meets Primary Coverage Criteria Or Is Covered For Contracts Without Primary Coverage Criteria
 
EGFR TESTING: Analysis of somatic variants in exons 18 through 21 (eg, G719X, L858R, T790M, S6781, L861Q) within the epidermal growth factor receptor (EGFR) meets member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness to predict treatment response to an EGFR tyrosine kinase inhibitor therapy (eg, erlotinib [Tarceva], gefitinib [Iressa], afatinib [Gilotrif], or osimertinib [Tagrisso]) in patients with advanced lung adenocarcinoma, large cell carcinoma, advanced squamous cell non-small-cell lung cancer, and non-small-cell lung cancer not otherwise specified.
 
Use of a circulating tumor DNA test (cobas® EGFR Mutation Test v2 ) to detect the above specific mutations of the EGFR gene meets primary coverage criteria when the volume of formalin-fixed paraffin-embedded tumor tissue (FFPET) available for testing is insufficient and the other above criteria are met.
 
KRAS Testing: Analysis of the KRAS gene for the presence of KRAS G12C mutation meets member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness in improving health outcomes to predict treatment response to sotorasib (Lumakras) in patients with locally advanced or metastatic non-small cell lung cancer (NSCLC) who have received one prior systemic treatment.
 
ALK TESTING: Analysis of somatic rearrangement mutations of the ALK gene meets member benefit certificate primary coverage criteria to predict treatment response to ALK inhibitor therapy (eg, crizotinib, alectinib, or brigatinib in patients with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded.
 
BRAF V600E TESTING: Analysis of BRAF V600E variant meets member benefit certificate primary coverage criteria to predict treatment response to BRAF or MEK-inhibitor therapy (eg, dabrafenib [Tafinlar] and trametinib [Mekinist]), in patients with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded.
 
ROS1 TESTING: Analysis of somatic rearrangement variants of the ROS1 gene meets member benefit certificate primary coverage criteria to predict treatment response to ALK inhibitor therapy (crizotinib [Xalkori]) in patients with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded.
 
NTRK GENE FUSION TESTING:  Analysis by immunohistochemistry (IHC) of NTRK gene fusions meets member benefit certificate primary coverage criteria to predict treatment response to entrectinib or larotrectinib in patients with advanced lung adenocarcinoma or in whom an adenocarcinoma component cannot be excluded.
 
RET REARRANGEMENT TESTING: Analysis of genetic alteration in the RET gene meets member benefit certificate primary coverage criteria to predict treatment response to pralsetinib (Gavreto) or selpercatinib (Retevmo) in patients with metastatic NSCLC.
 
MET EXON 14 SKIPPING ALTERATION: Analysis of genetic alteration that leads to MET exon 14 skipping gene meets member benefit certificate primary coverage criteria to predict treatment response to capmatinib (Tabrecta) in patients with metastatic NSCLC.
 
PD-L1 TESTING: PD-L1 testing gene meets member benefit certificate primary coverage criteria to predict treatment response to atezolizumab (Tecentriq), nivolumab (Opdivo) in combination with ipilimumab (Yervoy) or pembrolizumab (Keytruda) in patients with metastatic NSCLC.
 
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
 
Testing for the following indications does not meet member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness in improving health outcomes in any clinical situations or other applications not described above for NSCLC:
 
    • Analysis of other EGFR variants within exons 22 to 24;
    • Analysis of somatic mutations of the KRAS gene as a technique to predict treatment nonresponse to anti-EGFR therapy with the tyrosine-kinase inhibitors (eg, erlotinib) and for the use of the anti-EGFR monoclonal antibody (eg, cetuximab);
    • Analysis of genetic alterations in the HER2 gene for targeted therapy in patients with NSCLC;
    • Testing for genetic alterations in the genes RET and MET for targeted therapy in patients with NSCLC;
    • Analysis of somatic rearrangement mutations of the ALK gene.
 
For members with contracts without primary coverage criteria testing for the following indications are considered investigational. Investigational services are specific contract exclusions in most member benefit certificates of coverage in any clinical situations or other applications not described above for NSCLC:
 
    • Analysis of other EGFR variants within exons 22 to 24;
    • Analysis of somatic mutations of the KRAS gene as a technique to predict treatment nonresponse to anti-EGFR therapy with the tyrosine-kinase inhibitors (eg, erlotinib) and for the use of the anti-EGFR monoclonal antibody (eg, cetuximab);
    • Testing for genetic alterations in the genes RET and MET for targeted therapy in patients with NSCLC;
    • Analysis of genetic alterations in the HER2 gene for targeted therapy in patients with NSCLC;  
    • Analysis of somatic rearrangement mutations of the ALK gene.  
 
 
Due to the length of the policy, criteria for dates of service prior to October 2021, are not online. If you would like a hardcopy print, please email: codespecificinquiry@arkbluecross.com

Rationale:
“Due to the detail of the rationale, the complete document is not online. If you would like a hardcopy print, please email: codespecificinquiry@arkbluecross.com”
 
The most recent literature search conducted for EGFR mutation testing was through December 2013, for KRAS mutation testing through January 2014, and for all remaining mutations addressed in this policy through September 4, 2014.
 
EGFR
Two publications (Lynch, 2004; Paez, 2004) demonstrated that the underlying molecular mechanism underpinning dramatic responses in these favorably prognostic groups appeared to be the presence of activating somatic mutations in the TK domain of the EGFR gene, notably small deletions in exon 19 and a point mutation in exon 21 (L858R, indicating substitution of leucine by arginine at codon position 858). These can be detected by direct sequencing or PCR technologies.
 
A TEC Assessment on this topic was first published in November 2007. The Assessment concluded that there was insufficient evidence to permit conclusions about the clinical validity or utility of EGFR mutation testing to predict erlotinib sensitivity or to guide treatment in patients with NSCLC. This Assessment was updated in 2010, with revised conclusions indicating that EGFR mutation testing has clinical utility in selecting or deselecting patients for treatment with erlotinib.
 
A 2013 meta-analysis (Lee, 2013) of 23 trials of erlotinib, gefitinib, and afatinib in patients with advanced NSCLC reported improved progression free survival (PFS) in EGFR mutation-positive patients treated with EGFR TKIs in the first- and second-line settings and for maintenance therapy. (Comparisons were with chemotherapy, chemotherapy and placebo, and placebo in the first-line, second-line, and maintenance therapy settings, respectively.) Among EGFR mutation-negative patients, PFS was improved with EGFR TKIs compared with placebo maintenance but not in the first- and second-line settings. Overall survival (OS) did not differ between treatment groups in either mutation-positive or mutation-negative patients.
Statistical heterogeneity was not reported for any outcome. The authors concluded that EGFR mutation testing is indicated to guide treatment selection in NSCLC patients.
 
Erlotinib
Thirteen publications provide data on EGFR mutations in tumor samples obtained from NSCLC patients in erlotinib treatment studies. Nine of these (Ahn, 2008; Amann, 2010; Felip, 2008; Miller, 2008; chneider, 2008; Eberhard, 2005; Giaccone, 2006; Jackson, 2007; Zhu, 2008) were nonconcurrent-prospective studies of treatment naïve and previously-treated patients who received erlotinib and were then tested for the presence or absence of mutations; 4 (see Table 1) were prospective 1-arm enrichment studies of mutation-positive or wild-type patients treated with erlotinib. In 3 studies of EGFR mutation-positive patients (Jackman. 2009; Rosell, 2009; Sun, 2011) objective radiologic response was 40% to 70%, median PFS was 8 to 14 months, and median OS was 16 to 29 months. In patients with wild-type tumors (Yoshioka, 2010) objective radiologic response was 3.3%, PFS was 2.1 months, and overall survival (OS) was 9.2 months.
 
Clinical Response in Prospective Studies of Erlotinib Therapy in Patients with EGFR
Gene Mutation-Positive Advanced NSCLCa      
 
Abbreviation explanations for information below:
CI: confidence interval; OS: overall survival; PFS: progression-free survival.
a All patients had stage IIIA/IV NSCLC.
 
EGFR mutation positive
  • Jackman et al (2009) Prospective 1-arm treatment EGFR-positive patients with erlotinib, chemotherapy naïve (Jackman, 2009); No. Mutated/No. Tested (%) 84 enrolled; Objective Radiologic Response (%) 70; Median PFS (95%) CI), mo 13; Median OS (95% CI), mo 28.7.
  • Rosell et al (2009) Prospective 1-arm treatment EGFR-positive patients with erlotinib in treatment failure and chemotherapy naïve (Rosell, 2009) ; No. Mutated/No. Tested (%); 350/2105 16.6 enrolled; Objective Radiologic Response (%) 70; Median PFS (95%) CI), mo 14 (11.3 to 16.7); Median OS ((%% CI), mo 27. (24.9 to 33.1).
  • Sun et al (2010) Prospective 1-arm treatment EGFR-positive patients with erlotinib in treatment failures (Sun. 2011); No. Mutated/No. Tested (%); 144-164 (32) enrolled; Objective Radiologic Response (%)40; Median PFS (95%) CI), mo 8; Median OS (95% CI), mo 15.8.
EGFR mutation negative (wild type)
  • Yoshioka et al (2010) Prospective 1-arm treatment EGFR wild-type (Yoshioka, 2010); No. Mutated/No. Tested (%); 30 enrolled; Objective Radiologic Response (%) 3.3; Median PFS (95%) CI), mo 2.1; Median OS (95% CI), mo 9.2.
 
 
In 2011, Zhou et al reported the results of a phase 3 prospective clinical trial of first-line treatment of
Chinese patients with EGFR mutation (exon 19 deletion or L858R)-positive NSCLC (87% adenocarcinoma) randomized to treatment with erlotinib (n=83) or standard chemotherapy (gemcitabine plus carboplatin, n=82) (Zhou, 2011). PFS was significantly longer in patients who received erlotinib (13.1 vs 4.5 months; hazard ratio [HR], 0.16; p<0.001). Patients treated with erlotinib experienced fewer grade 3 and 4 toxic effects and more clinically relevant improvements in quality of life (Chen, 2013) than those who received chemotherapy. These results were duplicated in a European population in the 2012 EURTAC trial (NCT00446225), a multicenter, open-label, randomized Phase 3 trial (Rosell, 2012). Adult patients with EGFR mutations (exon 19 deletion or L858R mutation in exon 21) with NSCLC were randomized. Eighty-six received erlotinib, and 87 received standard chemotherapy. A planned interim analysis showed that the primary end point had been met. At the time the study was halted (Jan 26, 2011), median PFS was 9.7 months (95% confidence interval [CI], 8.4 to 12.3) vs 5.2 months (95% CI, 4.5 to 5.8) in the erlotinib and standard chemotherapy groups, respectively (HR=0.37; 95% CI, 0.25 to 0.54; p<0.001). Six percent of patients receiving erlotinib had treatment-related severe adverse events compared with 20% of those receiving a standard chemotherapy regimen.
 
In 2011, Petrelli et al (Petrelli, 2012) reported a meta-analysis of 13 randomized trials of 1260 patients with EGFR mutated NSCLC who received TKIs for first-line, second-line, or maintenance therapy, and compared outcomes with standard therapy. Overall, they noted that in patients, use of EGFR TKIs increased the chance of obtaining an objective response almost 2-fold when compared with chemotherapy. Response rates were 70% vs 33% in first-line trials and 47% vs 28.5% in second-line trials. Tyrosine kinase inhibitors reduced the hazard of progression by 70% in all trials and by 65% in first-line trials; however, they did not improve overall survival.
 
In a 2010 pooled analysis of patients with EGFR mutations (most commonly exon 19 deletions and
L858R substitution mutations in exon 21), median PFS was 13.2 months in patients treated with erlotinib and 5.9 months in patients treated with standard chemotherapy (p<0.001) (Paz-Ares, 2010). Patients with EGFR mutations appear to be ideal candidates for treatment with erlotinib. Identification of patients likely to respond or fail to respond to erlotinib leads to tailored choices of treatment likely to result in predictable and desirable outcomes.
 
Nine other studies totaling 630 patients have compared outcomes in EGFR mutation-positive and EGFR wild-type patients who were treated with erlotinib (see Table 2).
  • Objective radiologic response rates ranged from 0% to 83% (median, 45%) in patients with EGFR mutation-positive tumors and from 0% to 18% (median, 5.5%) in patients with wild-type tumors. All 5 studies that statistically evaluated results demonstrated statistically significant increases in objective radiologic response among patients with EGFR mutation-positive tumors.  
  • Progression free survival ranged from 6.8 to 13.1 months (median, 12.5) in patients with EGFR mutation-positive tumors and from 1.4 to 5 months (median, 2.5) in patients with wild-type tumors. In all studies in which these data were reported, patients with EGFR mutation-positive tumors showed a trend or a statistically significant increase in PFS.
  • Overall survival ranged from 10 to 35 months (median, 21) in patients with EGFR mutation positive tumors and from 3 to 12 months (median, 8.1) in patients with wild-type tumors. In all cases in which these data were reported, EGFR mutation-positive tumors showed a trend or a statistically significant increase in overall survival.
 
Outcomes in Patients According to EGFR Mutation Status in Response to Treatment With
Erlotinib (9 studies of 630 patients)
 
Abbreviation explanations for information below:
OS: overall survival; PFS: progression-free survival.
 
  • EGFR mutation-positive patients; overall radiologic response (range) 45% (0-83); medial PSF (range), mo 12.5 (6.8 – 13.1); medial OS (range), mo 21 (10-35).
  • Wild-type patients; overall radiologic response (range) 5.5% (0-18); medial PSF (range), mo 2.5 (1.4-5); medial OS (range), mo 8.1 (3-12).
  • Untested patients (intent to treat) – FDA label; overall radiologic response (range)not reported; medial PSF (range), mo 2.8; medial OS (range), mo12.
 
In a 2013 randomized controlled trial (RCT), Garassino et al in Italy compared the efficacy of erlotinib and docetaxel as second-line therapy in 219 EGFR wild-type patients with metastatic NSCLC who had received previous platinum-based therapy (Garassino, 2013). Most patients (69%) had adenocarcinoma; 25% had squamous cell carcinoma (SCC). With a median follow-up of 33 months, median PFS was 2.9 months with docetaxel and 2.4 months with erlotinib (adjusted HR=0.71; 95% CI, 0.53 to 0.95; p=0.02). Median overall survival was 8.2 months with docetaxel and 5.4 months with erlotinib (adjusted HR=0.73; 95% CI, 0.53 to 1.00; p=0.05). Grade 3 or higher skin adverse events occurred in 14% of the erlotinib group and did not occur in the docetaxel group. Grade 3 or higher neutropenia occurred only in the docetaxel group (20%). As stated in an accompanying editorial, “[T]he efficacy of EGFR tyrosine kinase inhibitors is very limited for second-line treatment of wild-type EGFR NSCLC” (Jassem, 2013). A 2013 meta-analysis of 3 trials in patients with wild-type EGFR reported improved overall survival with erlotinib treatment in second and third line and maintenance settings (Jazieh, 2013).  However, 75% of patients in the control arms in this analysis received placebo.
 
EGFR mutations may provide prognostic information (about disease recurrence and survival), as well as predictive information (about treatment response). In a 2005 study by Eberhard et al, improved outcomes were observed for EGFR mutation-positive patients compared with wild-type patients regardless of treatment (standard chemotherapy or standard chemotherapy plus erlotinib). Objective radiologic response was 38% versus 23% (p=0.01), median time to progression was 8 months versus 5 months (p<0.001), and median OS was not reached versus 10 months (p<0.001).
 
Afatinib
Unlike erlotinib (and gefitinib) that selectively inhibit EGFR, afatinib inhibits not only EGFR but also human epidermal growth factor receptor 2 (HER2) and HER4 and may have activity in patients with acquired resistance to TKIs (who often harbor a T790M mutation [substitution of threonine by methionine at codon 790] in EGFR exon 20). The efficacy and safety of afatinib was evaluated in the LUX-Lung series of studies.
 
LUX-Lung 3 was an RCT in 345 patients with stage IIIB or IV, EGFR mutation-positive, lung adenocarcinoma who were previously untreated for advanced disease (Sequist, 2013). Seventy-two percent of patients were Asian, 26% were white, and 90% (308 patients) had common EGFR mutations (exon 19 deletion or L858R substitution mutation in exon 21). Patients received either afatinib or chemotherapy (cisplatin plus pemetrexed). In stratified analysis of patients with common EGFR mutations, median PFS was 13.6 months for the afatinib group and 6.9 months for the chemotherapy group (HR=0.47; 95% CI, 0.34 to 0.65; p=0.001). Median PFS for the 10% of patients who had other EGFR mutations was not reported, but median PFS for the entire patient sample was 11.1 months in the afatinib group and 6.9 months in the chemotherapy group (HR=0.58; 95% CI, 0.43 to 0.78; p=0.001). Incidence of objective response in the entire patient sample was 56% in the afatinib group and 23% in the chemotherapy group (p=0.001). With a median follow-up of 16.4 months, median OS was not reached in any group; preliminary analysis indicated no difference in OS between the 2 treatment groups in the entire patient sample (HR=1.12; 95% CI, 0.73 to 1.73; p=0.60). Patients in the afatinib group reported greater improvements in dyspnea, cough, and global health status/quality of life than those in the chemotherapy group (Yang, 2013). Grade 3 or higher diarrhea, rash, and paronychia (nail infection) occurred in 14%, 16%, and 11% of afatinib-treated patients respectively, and in no patients in the chemotherapy group. Grade 3 or higher mucositis (primarily stomatitis) occurred in 9% of the afatinib group and 0.9% of the chemotherapy group (Sequist, 2013).
 
Three other published LUX-Lung studies evaluated patients with stage IIIB or IV lung adenocarcinomas who were previously treated for advanced disease, but each had design flaws that limit the interpretation of results.
 
  • LUX-Lung 2 was a single-arm study of afatinib in 129 patients (87% Asian, 12% white) with EGFR mutation-positive disease (Yang, 2012). Patients had been treated with previous chemotherapy but not with EGFR-targeted therapy; approximately half of patients (enrolled after a protocol amendment) were chemotherapy-naïve. Objective responses (primarily partial responses) were observed in 66% of 106 patients with common EGFR mutations (exon 19 deletion or L858R) and in 39% of 23 patients with other EGFR mutations. Median PFS was 13.7 months in patients with common EGFR mutations and 3.7 months in patients with other EGFR mutations (p values not reported). Results for mutation-negative patients were not reported.
  • LUX-Lung 1 and LUX-Lung 4 enrolled patients who had progressed on previous erlotinib, gefitinib, or both for advanced disease. Neither study prospectively genotyped patients. In the LUX-Lung 1 double-blind RCT(Miller, 2012), 96 of 585 enrolled patients (66% Asian, 33% white) were EGFR mutation-positive (76 common EGFR mutation-positive). In this group, median PFS was 3.3 months in the afatinib group and 1.0 month in the placebo group (HR=0.51; 95% CI, 0.31 to 0.85; p=0.009). In 45 mutation-negative patients, median PFS was 2.8 months in the afatinib group and 1.8 months in the placebo group, a statistically nonsignificant difference (p=0.22), possibly due to small group sizes. LUX-Lung 4 was a single-arm study of afatinib in 62 Japanese patients (Katakami, 2013). Objective responses occurred in 2 of 36 patients with common EGFR mutations (5%) and in none of 8 patients with other EGFR mutations (p>0.05).
 
EGFR Mutation Frequency
In 2009, Rosell et al reported EGFR mutations in 16.6% of the overall patient sample but noted an increased prevalence in women (69.7%), patients who never smoked (66.6%), and patients with adenocarcinomas (80.9%). Based on these findings, Rosell et al recommended EGFR mutation screening in women with lung cancer with nonsquamous cell tumors who have never smoked. Other reports on the mutation frequencies have found higher prevalences among East Asians when compared with other ethnicities (38% vs 15%, respectively) (Rosell, 2009). Although there is a greater proportion of EGFR mutations in these special populations (women, never smokers, patients with adenocarcinoma, and/or Asians), many patients without these selected demographics still exhibit EGFR mutations and would benefit from erlotinib treatment.
 
In a comprehensive analysis of 14 studies involving 2880 patients, Mitsudomi et al (Mitsudomi, 2006), reported EGFR mutations in 10% of men, 7% of non-Asian patients, 7% of current or former smokers, and 2% of patients with nonadenocarcinoma histologies. Although histology appeared to be the strongest discriminator, results varied across studies; for example, Eberhard et al (Eberhard, 2005) observed EGFR mutations in 6.4% of patients with squamous cell carcinomas (SCCs) and Rosell et al (Rosell, 2009) in 11.5% of patients with large cell carcinomas. (Both of these studies had small sample sizes.)
 
For patients with SCC, current guidelines from the National Comprehensive Cancer Network (NCCN) indicate that the low incidence of EGFR mutations in SCC “does not justify routine testing of all tumor specimens.” This conclusion is based on the Sanger Institute’s Catalogue of Somatic Mutations in Cancer
(COSMIC) (Forbes, 2012), that reported an observed EGFR mutation incidence of 2.7% in patients with SCC with an upper confidence limit for the true incidence of 3.6%. NCCN guidelines recommend consideration of mutation testing in never smokers with SCC or when biopsy specimens are small and histology is mixed. This recommendation was based on a case series of 13 patients with squamous or pseudosquamous histology (Paik, 2012). However, 7 patients (54%) were subsequently determined to have adenocarcinoma histology. All 6 remaining patients were never smokers, and all 6 had an exon 19 deletion or L858R substitution mutation in EGFR.
 
In 2013, the College of American Pathologists, International Association for the Study of Lung Cancer, and Association for Molecular Pathology published joint evidence-based guidelines for molecular testing to select EGFR TKI therapy in patients with lung cancer (Lindeman, 2013). An EGFR mutation incidence of 0% to 5% in patients with SCC was reported. Recommendations for EGFR mutation testing in patients with SCC depend on tumor sample availability:
 
  • For fully excised lung cancer specimens, EGFR testing is not recommended when an adenocarcinoma component is lacking, eg, tumors with pure squamous cell histology with no immunohistochemistry evidence of adenocarcinoma differentiation (eg, thyroid transcription factor 1 [TTF-1] or mucin positive). (Evidence grade A, excellent quality evidence)  
  • When lung cancer specimens are limited (eg, biopsy, cytology) and an adenocarcinoma component cannot be completely excluded, EGFR testing may be performed in cases showing squamous cell histology; clinical criteria (eg, lack of smoking history) may be useful to select a subset of these samples for testing. (Evidence grade A, excellent quality evidence)
 
Two studies may support the potential value of EGFR mutation testing in patients with SCC, particularly in Asian populations. However, similar studies have not been reported in non-Asian populations or in populations treated with erlotinib. A 2009 study by Park et al (Park, 2009) of preselected Korean patients treated with gefitinib reported EGFR mutations in 3 of 20 male smokers with SCC (15%), a patient subgroup expected to have a low prevalence of EGFR mutations based on demographics. Clinical response was observed in 2 of 3 mutation-positive patients and 1 of 17 wild-type patients; median PFS was 5.8 months in patients with mutated EGFR and 2.4 months in the wild-type group (p=0.07). In vivo analyses by Dobashi et al (Dobashi, 2011) showed that in Japanese patients with both adenocarcinomas and SCCs, EGFR mutations were associated with downstream phosphorylation of EGFR and constitutive activation of the EGFR pathway.
 
In contrast, Fang et al (2013) reported EGFR mutations (all L858R) in 2% (3 patients) of 146 consecutively treated Chinese patients with early stage SCC (Park, 2009) In a separate cohort of 63 Chinese patients with SCC who received erlotinib or gefitinib as second- or third-line treatment (63% never smokers, 21% women), EGFR mutation prevalence (all exon 19 deletion or L858R) was 23.8%. Objective response occurred in 26.7% of 15 EGFR mutation-positive and 2.1% of 48 mutation-negative patients (p=0.002). Median PFS was 3.9 months and 1.9 months, respectively (p=0.19). Based on these findings, the authors concluded that routine EGFR mutation testing of all SCC specimens is not justified.
 
EGFR Mutation Testing
Gene sequencing is generally considered an analytic gold standard. In 2010, the Canadian Agency for
Drugs and Technologies in Health published a rapid response report on EGFR mutation analysis. Based on 11 observational studies, the report authors concluded that PCR-based approaches identify EGFR mutations with a sensitivity equivalent to that of direct sequencing.
 
Ongoing and Unpublished Clinical Trials
A search of ClinicalTrials.gov in September 2014 found several phase 3 trials in patients with NSCLC and
EGFR mutations, assessing several different TKIs and clinical scenarios, including a TKI versus chemotherapy, a TKI +/- chemotherapy, use of TKIs in 1st versus subsequent lines of therapy and use of TKIs in the neoadjuvant setting.
 
Several randomized controlled trials, nonconcurrent prospective studies, and single-arm enrichment studies demonstrate that detection of epidermal growth factor receptor (EGFR) gene mutations identifies patients with non-small-cell lung cancer (NSCLC) who are likely to benefit from erlotinib or afatinib therapy and who are therefore ideal candidates for treatment with these drugs. These observations have been made in populations of patients with primarily adenocarcinomas. Currently, there is no little evidence to indicate that EGFR mutation testing can guide treatment selection in patients with squamous cell histology.
 
Patients who are found to have wild-type tumors are unlikely to respond to erlotinib or afatinib. These patients should be considered candidates for alternative therapies.
 
EGFR mutational analysis may be considered medically necessary to predict treatment response to erlotinib or afatinib in patients with advanced NSCLC; however, EGFR mutational analysis is investigational in patients with NSCLC of squamous cell type.
 
KRAS
 
KRAS and EGFR Tyrosine Kinase Inhibitors
Data on the role of KRAS mutations in non-small-cell lung cancer (NSCLC) and response to erlotinib are available from post hoc analyses of 2 phase 3 trials of TKIs in patients with wild-type (nonmutated) versus KRAS-mutated lung tumors; phase 2 trials; a large prospective study; retrospective single-arm studies; and 2 meta-analyses.
 
Pao et al (2005) were the first to suggest that patients with KRAS-mutated lung tumors were nonresponsive to treatment with epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors
(TKIs) (Pao, 2005). Thirty-six patients with bronchioloalveolar carcinoma underwent KRAS mutation analysis; 9 (25%) were found to harbor KRAS mutations. Response was by a single radiologist, who was blinded to patient outcome, using RECIST (Response Evaluation Criteria in Solid Tumors). None of 9 patients with KRAS-mutated tumors responded to erlotinib (p=0.553).
 
Zhu et al (2008) performed a post hoc subgroup analysis of KRAS mutations in patients with advanced
NSCLC who had failed standard chemotherapy and had been previously randomized to receive erlotinib or placebo. The original phase 3 trial (National Cancer Institute of Canada Clinical Trials Group Study
BR.21; 2005) was the first to demonstrate a significant survival advantage with the use of an EGFR TKI in previously treated NSCLC patients (Shepherd, 2005). In post hoc analysis, 206 (28%) of the original 731 tumors were tested for KRAS mutations, which were identified in 30 patients (15%). Among the 206 tested patients, 118 (57%) were assessable for response to erlotinib. Of 98 patients with wild-type KRAS, 10 (10.2%) responded to erlotinib; of 20 patients with mutated KRAS, 1 patient (5.0%) responded (hazard ratio [HR] [erlotinib vs placebo] in patients with mutated KRAS, 1.67; 95% confidence interval [CI], 0.62 to 4.50;
p=0.31]; HR in wild-type patients, 0.69; 95% CI, 0.49 to 0.97; p=0.03). In Cox regression, the interaction between KRAS mutation status and treatment was not statistically significant (p=0.09).
 
Eberhard et al (2005) performed a post hoc subgroup analysis of KRAS mutations in previously untreated patients with advanced NSCLC who had been randomly assigned to receive chemotherapy with or without erlotinib (Eberhard, 2005). The original phase 3 trial (TRIBUTE; 2005) randomly assigned patients to carboplatin plus paclitaxel either with or without erlotinib (Herbst, 2005). Of the original 1079 patients, tumor DNA from 274 patients (25%) was sequenced for KRAS mutations. Baseline demographics between patients with available tumor DNA and those without were balanced. KRAS mutations were detected in 55 of 274 patients (21%). Response rate for patients with wild-type KRAS was 26%, regardless of treatment received. In patients with KRAS-mutated tumors, response rate was 8% for those receiving chemotherapy with erlotinib and 23% for those receiving chemotherapy alone (p=0.16; 95% CI for difference: -5% to 35%); median overall survival (OS) was 4.4 months (95% CI, 3.4 to 12.9) in patients who received erlotinib and 13.5 months (95% CI, 11.1 to 15.9) in those who received chemotherapy alone (p=0.019).
 
In a 2007 a phase 2, multicenter, open-label study, Jackman et al evaluated treatment response to erlotinib in chemotherapy-naive patients 70 years of age or older who had advanced NSCLC (Jackman, 2007). Of 80 patients eligible for treatment, 41 (51%) had KRAS mutation analysis; 6 patients (15%) were mutation-positive, none of whom responded to erlotinib. Five (14%) of 35 patients with wild-type KRAS had a partial response.
 
In a 2008 phase 2 trial, Miller et al assessed response to erlotinib in 101 patients with lung bronchioloalveolar carcinoma (n=12) or adenocarcinoma, bronchioloalveolar subtype (n=89), according to
KRAS mutational status (Miller, 2008).  Eighteen patients (18%) had KRAS-mutated tumors, and none of them responded to erlotinib (95% CI, 0%-19%; p<0.01). In patients without a KRAS mutation, response rate was 32%. Median OS in patients with KRAS-mutated tumor was 13 months versus 21 months in patients with KRAS wild-type tumor (p=0.30).
 
In a 2006 phase 2 trial, Giaccone et al studied response to erlotinib in 53 chemotherapy-naive patients with advanced NSCLC (Giaccone, 2006). Histologic material was available to assess KRAS mutational status from 29 patients, 10 of whom (34%) had mutations. All 10 were nonresponders to erlotinib (p=0.125).
 
In 2009, Boldrini et al reported on the association between KRAS and EGFR mutation status and several clinical variables in 411 patients with lung adenocarcinoma, and presented a subgroup analysis of tumor response in patients treated with erlotinib or gefitinib (Boldrini, 2009).  KRAS mutations were observed in 17.9% of all patients. The subset analysis comprised 21 women with stage IV disease who received a TKI as second or third-line therapy and were assessed for radiographic tumor response using RECIST. Mean age of this subpopulation at the time of diagnosis was 60.8 years (range 40-86). Nineteen (90%) of 21 women were KRAS wild-type, and of those, 8 (42%) showed partial response, 4 (21%) had stable disease, and 7 (37%) had progressive disease. Two patients with KRAS mutations had progressive disease.
 
Schneider et al (2008) reported on the relationship between clinical benefit and putative tumor markers in a subgroup of patients participating in a global open-label, single-arm study of erlotinib in advanced
NSCLC, involving 7043 patients in 52 countries (the TRUST study) (Schneider, 2008). The subgroup in this publication was from German centers and comprised 311 patients with stage IIIB/IV disease who were treated with erlotinib because they had failed or were not medically suitable for standard first-line chemotherapy. Tumor response was assessed using RECIST. Seventeen patients (15%) had KRAS mutations, and none had a response to erlotinib; 2 patients had stable disease. The impact of KRAS mutation status on OS (p=0.06) and progression-free survival (PFS; p not reported) was of borderline statistical significance. The authors concluded that current data did not support selection of patients for treatment with erlotinib on the basis of tumor molecular characteristics and that further studies were needed to determine definitively whether patients with KRAS mutations can derive survival benefit from erlotinib.
 
Two meta-analyses on the relationship between KRAS mutations and response to EGFR TKI therapy are outlined below. Data were insufficient to make a determination about an association between KRAS mutation status and PFS or OS in these meta-analyses.
 
Linardou et al (2008) performed a meta-analysis of 17 studies with 1008 patients, 165 (16.4%) of whom had a KRAS mutation (Linardou, 2008). Eligible studies reported response (complete or partial) stratified by KRAS mutational status. Primary end points were sensitivity and specificity of KRAS testing, defined as KRAS mutation carriers showing no response to erlotinib (stable disease or progressive disease) and KRAS wild-type patients showing a response, respectively. Sensitivity and specificity were assessed overall and in subgroups defined by TKI received (gefitinib and/or erlotinib), response criteria (RECIST or World Health Organization), possible selection bias, and previous chemotherapy, if any. There was no significant difference in sensitivity or specificity across subgroups. The presence of a KRAS mutation was associated with a lack of response to TKIs (sensitivity: 0.21; 95% CI, 0.16 to 0.28; specificity: 0.94; 95%\ CI, 0.89 to0.97; positive likelihood ratio: 3.52; negative likelihood ratio: 0.84). (For the analysis, likelihood ratios were calculated by using pooled estimates for sensitivity and specificity.) The authors concluded that KRAS mutations conferred a high level of resistance to anti-EGFR therapies; however, this conclusion is tentative due to limitations of the study, such as lack of individual patient data. Prospective validation is needed. Furthermore, incomplete reporting of survival data precluded meaningful assessment of the effect of KRAS mutation on survival. Other limitations included heterogeneity of response end points, treatment regimens, and patient selection criteria, and the retrospective design of included studies.
 
Mao et al (2010) performed a meta-analysis of 22 studies in 1470 patients with NSCLC (1335 [91%] evaluable for response), 231 (17%) of whom had KRAS mutations (Mao, 2010). Studies were heterogeneous in patient populations (smoking history, tumor histology, stage, ethnicity, and treatment received) and response criteria. The primary end point was objective response rate, defined as the sum of complete and partial response. Objective response rates for patients with mutated KRAS and wild-type KRAS were 3% and 26%, respectively. Incomplete reporting of survival data precluded meaningful assessment of th effect of KRAS status on survival in NSCLC patients treated with EGFR TKIs. Data for PFS and OS stratified by KRAS status were available in 8 studies. Median PFS in KRAS-mutated and wild-type patients was 3.0 months and 3.9 months, respectively. Median OS in KRAS-mutated and wild-type patients was 4.7 months and 10.7 months, respectively. However, only 2 studies presented hazard ratios with 95% confidence intervals for PFS and OS, and therefore, pooled analysis to derive an overall HR was not performed.
 
Guan et al (2013) reported on 1935 consecutive patients with NSCLC who were treated at a single institution in China (Guan, 2013) Patients with mutated KRAS were random matched on tumor, node, metastasis (TNM) stage, time of first visit within 1 year, and histology, to both EGFR mutation-positive and KRAS/EGFR wild-type patients. Seventy patients (4%) received EGFR TKI therapy. In this group, median PFS was 11.8 and 2.0 months in patients with EGFR and KRAS mutations, respectively, and 1.9 months in wild-type patients; in comparison with wild-type patients, PFS was statistically longer in patients with EGFR mutations (p<0.001) but no different in patients with KRAS mutations (p=0.48). The authors observed that “the presence of an EGFR mutation, but not a KRAS mutation, was predictive of responsiveness to EGFR TKI treatment.”
 
Fiala et al (2013) reported on a retrospective analysis of patients with squamous cell NSCLC who underwent EGFR, KRAS, and PIK3CA (phosphatidylinositide-3-kinase catalytic subunit-alpha) mutation testing (Fiala, 2013). Of 215 patients tested, 16 (7.4%) had mutated KRAS. Of 174 tested patients who were treated with an EGFR TKI (erlotinib or gefitinib), median PFS in 14 KRAS-mutated patients was 1.3 months versus 2.0 months in KRAS wild-type patients (n=160 [92%]); the difference was not statistically significant (Kaplan-Meier [KM] log-rank test, p=0.120). Median OS in this treated group was 5.7 months in
KRAS-mutated patients versus 8.2 months in KRAS wild-type patients, a statistically significant difference
(KM log-rank test, p=0.039). The authors concluded that KRAS mutation status may have a negative prognostic role but a predictive role was not confirmed. “Patients with squamous cell NSCLC harboring these mutations could benefit from targeted treatment and should not be excluded from treatment with EGFR TKIs.”
 
Two reviews published in 2013 concluded that, in comparison with KRAS mutation testing, EGFR mutation status is the preferred predictive marker for response to EGFR TKIs in patients with NSCLC.59,60
 
.KRAS and Anti-EGFR Monoclonal Antibodies
Two phase 3 trials, BMS-099 and FLEX, investigated platinum-based chemotherapy with and without cetuximab in the first-line setting for advanced NSCLC. Subsequently, investigations of KRAS mutation status and cetuximab treatment were performed for both trials.
 
In the multicenter phase 3 BMS099 trial (2010), 676 chemotherapy-naive patients with stage IIIB/IV
NSCLC were assigned to taxane and carboplatin with or without cetuximab (Lynch, 2010). The primary end point was
PFS; secondary end points were overall response rate, OS, quality of life, and safety. The addition of cetuximab did not significantly improve PFS; however, there was a statistically significant improvement in overall response rate in the cetuximab group. There was a trend in OS favoring cetuximab; however, this was not statistically significant. A post hoc correlative analysis of this trial was conducted to identify molecular markers for the selection of patients most likely to benefit from cetuximab (Khambata-Ford, 2010).  Of the original 676 enrolled patients, 202 (29.9%) had tumor samples available for KRAS testing. KRAS mutations were present in 35 patients (17%). Among patients with wild-type KRAS, OS was similar between the cetuximab-containing arm (n=85) and the chemotherapy alone arm (n=82) (HR=0.93; 95% CI, 0.67 to 1.30; p=0.68; median survival, 9.7 and 9.9 months, respectively). Among patients with KRAS mutations, OS was similar between the cetuximab-containing arm (n=13) and the chemotherapy-alone arm (n=22) (HR=0.91; 95% CI, 0.45 to 2.07; p=0.93; median survival, 16.8 and 10.8 months, respectively). Overall, the study showed no significant treatment-specific interactions between the presence of KRAS mutations and outcomes evaluated; treatment differences favoring the addition of cetuximab in the KRAS-mutated subgroup were consistent with those observed in the wild-type KRAS subgroup and in the overall study population. The authors concluded that the results do not support an association between KRAS mutation and lack of cetuximab benefit similar to that observed in patients with KRAS-mutated metastatic colorectal cancer. However, results should be interpreted with caution due to small subgroup sample sizes and retrospective nature of the analysis.
 
In the open-label, randomized, phase 3 FLEX trial, 1125 chemotherapy-naive patients with stage III/IV,
NSCLC were randomly assigned to receive either chemotherapy (cisplatin and vinorelbine) plus cetuximab (n=557) or chemotherapy alone (n=568) (Pirker, 2009P). The primary end point was OS. Patients who received chemotherapy plus cetuximab survived longer than those who received chemotherapy only (median OS, 11.3 months vs 10.1 months, respectively; HR for death, 0.87; 95% CI, 0.76 to 1.00;
p=0.04). Subsequently, KRAS mutation testing was performed on archival tumor tissue of 395 (35%) of 1125 patients (O’Byrne, 2011). KRAS mutations were detected in 75 tumors (19%). Among patients with mutated KRAS, OS in the cetuximab-containing (n=38) and chemotherapy-alone arms (n=37) was similar (median OS, 8.9 months vs 11.1 months, respectively; HR=1.00; 95% CI, 0.60 to 1.66; p=1.0). Among patients with wild-type KRAS, OS in the cetuximab-containing (n=161) and chemotherapy-alone arms (n=159) was similar (median OS, 11.4 months vs 10.3 months, respectively; HR=0.96; 95% CI, 0.75 to 1.23; p=0.74). PFS also was similar in cetuximab-containing and chemotherapy-alone arms in patients with mutated (HR=0.97; 95% CI, 0.76 to 1.24) and wild-type (HR=0.84; 95% CI, 0.50 to 1.40) KRAS. Response rates in the cetuximab-containing arm in patients with KRAS-mutated and wild-type tumors were 36.8% and 37.3%, respectively (p=0.96). Overall, there was no indication that KRAS mutation status was predictive of cetuximab effect in NSCLC.
 
Ongoing and Unpublished Clinical Trials
A phase 3 trial is currently recruiting patients to assess progression-free survival and secondarily OS with
dacomitinib, another selective tyrosine kinase inhibitor, compared with erlotinib for the treatment of advanced NSCLC in patients who have received one or more prior anti-cancer therapies. (NCT01360554)
KRAS mutation status will be collected at baseline. Estimated enrollment is 800, with an estimated study completion date of September 2014.
 
Section summary
KRAS and EGFR Tyrosine Kinase Inhibitors
Data on the role of KRAS mutations in NSCLC and response to erlotinib are available from post hoc analysis of 2 phase 3 trials that compared TKI efficacy in patients with wild-type (nonmutated) versus
KRAS-mutated lung tumors; phase 2 trials; a large prospective study; retrospective single-arm studies; and 2 meta-analyses. Although studies have shown that KRAS mutations in patients with NSCLC confer a high level of resistance to TKIs, data are insufficient to assess any association between KRAS mutation status and survival in these patients.
 
KRAS and Anti-EGFR Monoclonal Antibodies
A lack of response to EGFR monoclonal antibodies has been established in metastatic colorectal cancer, and use of these drugs is largely restricted to patients with wild-type KRAS. The expectation that KRAS mutation status also would be an important predictive marker for cetuximab response in NSCLC has not been shown. In 2 randomized trials with post hoc analyses of KRAS mutation status and use of cetuximab with chemotherapy, KRAS mutations did not identify patients who would not benefit from anti-
EGFR antibodies, as outcomes with cetuximab were similar regardless of KRAS mutation status.
 
ROS
Bergethon and colleagues conducted a retrospective analysis of the clinical characteristics and treatment outcomes of patients with NSCLC with a ROS1 rearrangement (Bergethon, 2012). The authors screened 1,073 patients from multiple institutions for ROS1 rearrangements using a FISH assay and correlated ROS1 status with clinical characteristics, overall survival, and when available, ALK rearrangement status. Clinical data were extracted from medical record review. In vitro studies with human NSCLC cell lines were also conducted to assess the responsiveness of cells with ROS1 rearrangements to crizotinib. Of the tumors that were screened, 18 (1.7%) had ROS1 rearrangements, and 31 (2.9%) had ALK rearrangements. All of the ROS1-positive tumors were adenocarcinomas. The patients with ROS1 rearrangements were significantly younger (median age 49.8 years) and more likely to be never-smokers, when compared to the ROS1-negative group (each p<.001). There was no survival difference observed between the ROS1- positive and negative groups. The in vitro studies showed evidence of sensitivity to crizotinib. Finally, the authors reported the clinical response of one patient in their study with a ROS1 rearrangement. The patient was enrolled as part of an expanded phase 1 cohort, in an open-label, multicenter trial of crizotinib. The patient had no EGFR mutation or ALK rearrangement. After treatment failure with erlotinib, the patient was treated with crizotinib and had near complete resolution of tumor without evidence of recurrence at 6 months.
 
Kim and colleagues reported clinical outcomes in 208 never-smokers with NSCLC adenocarcinoma, according to ROS1-rearrangement status (Kim, 2013). ALK rearrangements and EGFR mutations were concurrently analyzed. The patients had clinical stages ranging from I-IV, but the majority were stage IV (41.3%). Of the 208 tumors, 3.4% (n=7) were ROS1 rearranged. ROS1 rearrangement was mutually exclusive from ALK rearrangement, but 1 of 7 ROS1-positive patients had a concurrent EGFR mutation.
Patients with ROS1 rearrangement had a higher objective response rate and longer median PFS on pemetrexed than those without a rearrangement. In patients with ROS1 rearrangement, PFS with EGFR-tyrosine kinase inhibitors was shorter than those patients without the rearrangement. None of the ROS1 positive patients received ALK inhibitors (eg, crizotinib), which is the proposed targeted therapy for patients with NSCLC and this genetic alteration.
 
RET
In a phase 2 prospective trial for patients with RET fusion-positive tumors, preliminary data on 3 patients treated with cabozantinib showed a partial response in 2 patients, and 1 with stable disease approaching
8 months (Drilon, 2013).
 
MET
A phase 2 trial of MET-positive NSCLC, in which patients were treated with an anti-MET antibody plus erlotinib, showed improved progression-free survival and overall survival (Sadiq, 2013).
 
BRAF
Rare case reports have documented a response to vemurafenib in patients with NSCLC and a BRAF mutation (Gautschi, 2012; Peters, 2013; Robinson, 2014).
 
HER2
Mazières and colleagues reported on a retrospective review of a consecutive series of patients with
NSCLC who were tested for a HER2 mutation, and the authors assessed clinicopathologic characteristics and patient outcomes according to mutation status (Mazieres, 2013). A HER2 mutation was identified in 65 of 3,800 (1.7%) patients, and was mutually exclusive of other driver mutations (EGFR, ALK and BRAF), with the exception of one case in which both a HER2 and KRAS mutation were identified. The patient population in which a HER2 mutation was found had a median age of 60 years (range 31 to 86), 69% were women, and 52% were never smokers. All of the tumors were adenocarcinomas, and 50% were stage IV (n=33). The patients with stage IV disease received conventional chemotherapy, and of these, 16 patients also received HER2-targeted therapy as additional lines of therapy (for a total of 22 individual anti-HER2 treatments that were evaluable). Four patients had progressive disease, 7 had disease stabilization, and 11 with partial response. PFS for patients with HER2 therapies was 5.1 months.
 
Summary
Over half of patients with non-small cell lung cancer (NSCLC) present with advanced and therefore incurable disease, and treatment in this setting has generally been with platinum-based chemotherapy.
More recently, the identification of specific, targetable oncogenic “driver” mutations in a subset of NSCLCs has resulted in a reclassification of lung tumors to include molecular subtypes, which are predominantly of adenocarcinoma histology.
 
In NSCLC, the first successful example of targeted therapy involved mutations in the epidermal growth factor receptor (EGFR) gene, in which lung tumors harboring specific activating mutations in the EGFR kinase domain showed a high sensitivity to EGFR tyrosine kinase inhibitors (TKIs). Phase 3 studies comparing EGFR-TKIs to chemotherapy in patients with EGFR-mutated NSCLC have shown that TKIs are superior to chemotherapy in terms of tumor response rate and progression free survival, with a reduction in toxicity and improvement in quality of life.
 
Currently, routine testing of NSCLC for EGFR mutations is recommended in patients with non-squamous
NSCLC, since TKIs are recommended if the tumor has an EGFR mutation.
 
Therefore, EGFR mutational analysis may be considered medically necessary to predict treatment response to erlotinib or afatinib in patients with advanced NSCLC; however, EGFR mutational analysis is investigational in patients with NSCLC of squamous cell type.
 
KRAS mutations may be prognostic in NSCLC and may predict a lack of response to TKIs, but the impact of testing for these mutations on clinical management is unknown. Studies have not shown that KRAS mutations identify a population that may benefit from the use of anti-EGFR monoclonal antibodies. Therefore, analysis of somatic mutations of the KRAS gene is considered investigational as a technique to predict treatment non-response to anti-EGFR therapy with the tyrosine-kinase inhibitors erlotinib and the anti-EGFR monoclonal antibody cetuximab in non-small-cell lung carcinoma.
 
Other, potentially targetable oncogenic mutations have been characterized in lung adenocarcinomas, including in the genes ROS, RET, MET, BRAF and HER2. The data on the use of targeted therapies in NSCLC with a mutation in one of these genes is preliminary, in that much of the demonstrated sensitivity of tumor to the various drugs has been in vitro or in animal studies, and published data on patient tumor response and survival outcomes are extremely limited, consisting of case reports and small case series. Therefore, testing for genetic alterations in the genes ROS, RET, MET, BRAF and HER2, for targeted therapy in patients with NSCLC, is considered investigational.
 
 
National Comprehensive Cancer Network (NCCN) Guidelines
NCCN guidelines for the treatment of NSCLC (version 4.2014) recommend the following:
 
EGFR mutation testing is recommended (category 1) in patients with non-squamous NSCLC (i.e. adenocarcinoma, large cell carcinoma) or in NSCLC not otherwise specified, because erlotinib or afatinib (category 1 for both) is recommended for patients who are positive for EGFR mutations.
Erlotinib is recommended as first-line therapy in patients with sensitizing EGFR mutations and should not be given as first-line therapy to patients negative for these EGFR mutations or with unknown EGFR status. Afatinib is recommended as first- or second-line therapy “for select patients with sensitizing EGFR mutations.” In patients with squamous cell carcinoma, EGFR mutation testing should be considered “especially in” never smokers; when histology is assessed using small biopsy specimens (rather than surgically resected samples); or when histology is mixed adenosquamous.
 
NCCN states that “KRAS mutations are associated with intrinsic TKI resistance, and KRAS gene sequencing could be useful for the selection of patients as candidates for TKI therapy”.
 
NCCN does not give specific recommendations for testing for genetic alterations in the genes ROS, RET,
MET, BRAF or HER2 in NSCLC, however, they state that the following targeted agents are now recommended for patients with specific genetic alterations: afatinib, cabozantinib, crizotinib, dabrafenib, erlotinib, gefitinib, trastuzumab and vemurafenib (category 2A).
 
American Society of Clinical Oncology Provisional Clinical Opinion
In 2011, the American Society of Clinical Oncology issued a provisional clinical opinion on EGFR mutation testing for patients with advanced NSCLC who are considering first-line EGFR tyrosine kinase inhibitor therapy (Keedy, 2011). The authors concluded that such patients who have not previously received chemotherapy or an EGFR tyrosine kinase inhibitor (TKI) should undergo EGFR mutation testing to determine whether chemotherapy or an EGFR TKI is appropriate first-line treatment.
 
College of American Pathologists Joint Guideline
In 2013, the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology published evidence-based guidelines for molecular testing to select patients with lung cancer for treatment with EGFR TKI therapy (Lindeman, 2013). Based on excellent quality evidence (category A), the guidelines recommend EGFR mutation testing in patients with lung adenocarcinoma regardless of clinical characteristics, such as smoking history. Guidelines for EGFR mutation testing in patients with SCC are reviewed in the Rationale section of the policy (see EGFR Mutation Frequency).
 
American College of Chest Physicians Guidelines
American College of Chest Physicians updated its evidence-based clinical practice guidelines on the treatment of stage IV NSCLC in 2013 (Socinski, 2013). Based on their review of the literature, guideline authors reported improved response rates, PFS, and toxicity profiles with first-line erlotinib or gefitinib compared with firstline platinum-based therapy in patients with EGFR mutations, especially exon 19 deletion and L858R. ACCP recommends “testing patients with NSCLC for EGFR mutations at the time of diagnosis whenever feasible, and treating with first-line EGFR TKIs if mutation-positive.”
 
2015 Update
This policy is being updated with a literature search using the MEDLINE database through April 2015. This update focuses on analysis of somatic rearrangement mutation of the ALK gene.
 
The accelerated approval of crizotinib by the FDA was based on phase 1 and 2 trials in which crizotinib showed marked antitumor activity in patients with ALK-positive advanced NSCLC, with objective response rates of 60% and PFS of 7 to 10 months (Camidge, 2012; Kwak, 2010). These results were confirmed in 2 subsequent phase 3 trials.
 
A phase 3, open-label trial randomized 347 patients with previously treated, locally advanced or metastatic ALK-positive lung cancer to oral crizotinib twice daily (n=173) or chemotherapy (n=174) every 3 weeks (Shaw, 2013). All patients had received one platinum-based chemotherapy regimen prior to the trial. The extent of disease was metastatic in 95% and 91% of patients in the crizotinib and chemotherapy groups, respectively, and tumor histology was adenocarcinoma in 95% and 94%, respectively. The primary endpoint was PFS. Patients in the chemotherapy group who experienced progressive disease were allowed to cross over to receive crizotinib as part of a separate study. Median PFS was 7.7 months in the crizotinib group versus 3.0 months in the chemotherapy group (HR for progression or death with crizotinib, 0.49; 95% CI 0.37 to 0.64; p<0.001). Partial response rates with crizotinib were 65% (95% CI, 58 to 72), versus 20% (95% CI, 14 to 26) with chemotherapy (p<0.001). Interim analysis of OS showed no significant improvement with crizotinib compared with chemotherapy (HR for death in the crizotinib group, 1.02; 95% CI, 0.68 to 1.54; p=0.54). Median follow-up for OS was 12.2 months and 12.1 months in the crizotinib and chemotherapy groups, respectively. Patients reported greater reductions in lung cancer symptoms and greater improvement in global quality of life with crizotinib compared with chemotherapy.
 
A phase 3, open-label trial compared crizotinib and chemotherapy in 343 previously untreated patients with ALK-positive advanced non-squamous NSCLC (Solomon, 2014). Patients were randomized to oral crizotinib twice daily or pemetrexed with either cisplatin or carboplatin every three weeks for up to 6 cycles. If there was disease progression for patients receiving chemotherapy, crossover to crizotinib was allowed. PFS was the primary end point. PFS was 10.9 months compared to 7.0 months for the group that received crizotinib versus chemotherapy, respectively (HR for progression or death with crizotinib, 0.45; 95% CI 0.35 to 0.60; p<0.001); objective response rates (complete and partial responses) were 74% and 45%, respectively (p<0.001). Median OS was not reached in either group; the probability of 1-year survival with crizotinib was 84% and with chemotherapy 79%. Crizotinib was associated with patient reported greater reduction in lung cancer symptoms and greater improvements in quality of life.
 
Crizotinib was granted accelerated approval by FDA in August 2011, for patients with locally advanced or metastatic NSCLC, based on objective response rates observed in 2, single-arm trials. Two subsequent phase 3 trials showed superior PFS and tumor response rates and improved quality of life in patients with crizotinib versus chemotherapy, in both previously untreated, ALK-positive advanced NSCLC and in patients who had received previous chemotherapy.
 
NCCN Guidelines
NCCN (v.4.2015) states that ALK rearrangement testing is recommended (category 1) in patients with non-squamous NSCLC (ie, adenocarcinoma, large cell carcinoma) or in NSCLC not otherwise specified, because crizotinib (category 1) is recommended for patients who are positive for ALK rearrangements. If ALK-positive status is discovered prior to first-line chemotherapy, give crizotinib (category 1), or if ALK rearrangement is discovered during first-line chemotherapy, interrupt or complete planned chemotherapy and start crizotinib. If there is progression on crizotinib, NCCN guidelines recommend multiple options (category 2A) including continuing crizotinib, switching to ceritinib, and considering local therapies depending upon symptoms.
  
2017 Update
A literature search conducted through September 2017 did not reveal any new information that would prompt a change in the coverage statement.  The key identified literature is summarized below.
 
Clinical Validity
EGFR-sensitizing and resistance variants can be detected by direct sequencing, or polymerase chain reaction (PCR) technologies, or next generation sequencing (NGS). Gene sequencing is considered an analytic criterion standard. In 2010, the Canadian Agency for Drugs and Technologies in Health published a rapid response report on EGFR mutation variant analysis (Mojoomdar, 2010).  Based on 11 observational studies, the report authors concluded that PCR-based approaches identify EGFR mutations variants with a sensitivity equivalent to that of direct sequencing
 
Four tests have been approved as companion diagnostics to detect EGFR-resistance variants (exon 19 deletions or exon 21 L858R substitutions) for at least one of the EGFR TKIs (afatinib, erlotinib, or gefitinib): the therascreen EGFR Rotor-Gene Q polymerase chain reaction (RGQ PCR) kit, cobas EGFR Mutation Test v1 and v2, Oncomine Dx Target Test. The cobas v2 test also is approved as a companion diagnostic to detect the T790M resistance variant to select patients for treatment with osimertinib.
 
The clinical validity of the therascreen RGQ PCR kit was demonstrated in a retrospective analysis of patients screened for a phase 3, open-label randomized controlled trial (RCT) comparing afatinib with chemotherapy in treatment-naive patients with stage IIIB or IV NSCLC, in which the EGFR variants for enrollment were determined using a Clinical Trial Assay (CTA) conducted at central laboratories (FDA, 2013/P120022b). The positive percent agreement (PPA) of therascreen vs CTA for detection of EGFR-sensitizing variants was 98% (95% confidence interval [CI], 95% to 99%) and negative percent agreement (NPA) was 97% (95%
CI, 94% to 99%). Overall, a statistically significant efficacy benefit for afatinib vs chemotherapy was
 
The clinical validity of the cobas EGFR Mutation Test v1 was demonstrated in a retrospective analysis of patients screened for a phase 3, open-label RCT comparing erlotinib with chemotherapy in treatment-naïve patients with advanced NSCLC. In this RCT, the EGFR variants for enrollment were determined with a CTA at a central laboratory using Sanger sequencing first for determination of EGFR variants status, followed by confirmatory testing for exon 19 deletions and exon 21 L858R variants (FDA, 2013/P120019b). The PPA of cobas vs CTA for detection of EGFR-sensitizing variants was 94% (95% CI, 89% to 97%) and NPA was -98% (95% CI, 95% to 99%). Overall, a statistically significant efficacy benefit for erlotinib vs chemotherapy was reported in the EGFR-positive patients as measured by the cobas EGFR Mutation Test v1 (HR=0.34; 95% CI, 0.21 to 0.54) that was similar to the efficacy in the overall population, which was EGFR-positive by the CTA (HR=0.34; 95% CI, 0.23 to 0.49). The cobas EGFR Mutation Test v2 expanded the indication for the use of the cobas EGFR Mutation Test to add the detection of the exon 20 (T790M) substitution variant in NSCLC patients for whom Tagrisso (osimertinib) treatment is indicated (FDA, v2 2015/P120019S007b). The clinical validity of the cobas EGFR Mutation Test v2 was demonstrated in retrospective analyses of patients enrolled in a phase 2, single-arm study of osimertinib for EGFR-sensitizing variant-positive metastatic NSCLC who had progressed following prior therapy with an approved EGFR TKI The osimertinib response rate in the patients identified as EGFR T790M+ by the cobas v2 test was 62% (95 % CI, 55% to 69%).
 
Similar results were reported in another phase 3 trial conducted in 364 Asian patients (Lux-Lung 6) comparing afatinib with gemcitabine plus cisplatin (Wu, 2014). PFS was 11.0 in the afatinib group and 5.6 months in the chemotherapy group (HR=0.28; 95% CI, 0.20 to 0.39) and the response rate was 67% and 23%.
 
Comparative Effectiveness of EGFR TKIs
As the previous sections have shown, erlotinib, gefitinib, and afatinib all have improved efficacy compared with chemotherapy in patients who have NSCLC and EGFR-sensitizing variants and are well tolerated. RCTs directly comparing the EGFR TKIs have been conducted.
 
Yang and colleagues published a systematic review and meta-analysis of studies comparing erlotinib, gefitinib, and afatinib (Yang, 2017). Eight randomized trials (N=2225 patients) and 82 cohort studies (N=15,396 patients) were included. However, results were not presented for trials vs cohort studies separately. Of the 8 RCTs identified, three were published in journals not available through PubMed, one was available as an abstract only, one was conducted in squamous cell NSCLC, and one did not require testing to confirm EGFR variant status for enrollment; these RCTs will not be discussed further (Park, 2016; Urata, 2016).
 
Two RCTs have compared gefitinib and erlotinib in patients with EGFR-sensitizing variants. Urata et al (2016) reported on a phase 3 RCT of 401 patients with EGFR variants randomized to gefitinib or erlotinib (Urata, 2016). The median PFS was 8.3 months (95% CI, 7.2 to 9.7 months) for patients receiving gefitinib and 10.0 months for those receiving erlotinib (95% CI, 8.5 to 11.2 months). Rash was more common with erlotinib (18.1% vs 2.2%) while both alanine aminotransferase elevation and aspartate aminotransferase elevation were more common with gefitinib (6.1% vs 2.2% and 13.0% vs 3.3%, respectively). Similarly, Yang and colleagues reported a median PFS of 13.0 for erlotinib and 10.4 months for gefitinib (HR=0.81; 95% CI, 0.62 to 1.05) in 256 patients with no differences in rates of grade 3 of 4 adverse events (Yang, 2017).
 
ALK GENE REARRANGEMENTS
Clinical Validity
Several methods are available to detect ALK gene rearrangements or the resulting fusion proteins in tumor specimens including fluorescence in situ hybridization (FISH), immunohistochemistry, reverse transcription polymerase chain reaction of cDNA (RT-PCR) and NGS. Two tests have been approved by the FDA as companion diagnostics to detect ALK rearrangements for treatment with crizotinib: the Vysis ALK Break Apart FISH Probe Kit and Ventana ALK (D5F3) CDx Assay.
 
The Vysis kit is a FISH-based assay. The clinical validity of the Vysis ALK Break Apart FISH Probe Kit was demonstrated in a retrospective analysis of patients screened for a phase 2, open-label single-arm study of crizotinib in patients with stage IIIB or IV NSCLC (FDA, 2011/p110012B). The response rate for crizotinib in 136 ALK-positive patients was 50% (95% CI, 42% to 59%) with a median duration of response of 42 weeks (range, 6-42). The response rate for 19 ALK-negative patients was 26% (95% CI, 9% to 51%).
 
The Ventana assay is an ICH-based assay. The clinical validity of the Ventana ALK (D5F3) CDx Assay was demonstrated in a retrospective analysis of patients screened for an open-label RCT of crizotinib vs platinum-doublet chemotherapy in patients with stage IIIB/IV NSCLC (FDA, 2015/PP140025B). The concordance between the Ventana and Vysis tests were calculated using patient samples analyzed at an independent, central laboratory. The PPA was 86.0% (95% CI, 80.2% to 90.4%) and the NPA was 96.3% (94.7% to 97.4%). Overall, in 343 patients who were ALK-positive by the Vysis assay, crizotinib was associated with longer PFS compared with chemotherapy (HR=0.45; 95% CI, 0.36 to 0.60). In the subset of 141 patients who were also ALK-positive by the Ventana assay, the results were similar (HR=0.40; 95% CI, 0.25 to 0.64). In the 25 patients that were ALK-positive by the Vysis assay and ALK-negative by the Ventana assay, the relative effect of crizotinib was not clear (HR=1.71; 95% CI, 0.43 to 6.79).
 
Other ALK Inhibitors
Three other ALK TKIs are FDA-approved but without separate companion diagnostics. Ceritinib has demonstrated superior efficacy concerning PFS when compared with chemotherapy in both the first-line and second-line (following crizotinib) settings in the ASCEND-4 and ASCEND-5 RCTs (Shaw, 2017; Soria, 2017). Alectinib was associated with response rates of approximately 50% in patients who had progressed on crizotinib in 2 phase 2 studies (Ou, 2016; Shaw, 2016). Alectinib has also shown superior efficacy and lower toxicity when compared with crizotinib in the first-line setting in the ALEX and J-ALEX phase 3 RCTs (Hida, 2017; Peters, 2017). Brigatinib has shown promise in early phase 1 and phase 2 studies with PFS of almost 13 months in patients with crizotinib-refractory disease (Gettinger, 2016; Kim, 2017).
 
ROS GENE REARRANGEMENTS
Clinical Validity
Several methods are available to detect ROS1 translocations including FISH, immunohistochemistry, and quantitative real-time reverse transcription-PCR (qRT-PCR) and some NGS panels. FISH is considered the standard method. The Oncomine Dx Target Test was FDA-approved in 2017 as a companion diagnostic to detect fusions in ROS1 to aid in selecting NSCLC patients for treatment with Xalkori (crizotinib). The Oncomine test is an NGS oncology panel that detects, among other variants, fusions in ROS1 from RNA isolated from formalin-fixed, paraffin-embedded (FFPE) tumor tissue samples (FDA, 2017/P160045b). The clinical validity of the detection of ROS1 rearrangements by the test was evaluated by retrospective analysis of FFPE NSCLC specimens obtained from patients enrolled in a ROS1 cohort from an ongoing single-arm, phase 1 safety, study of crizotinib in patients with advanced cancer. The ROS1 fusion status was determined by a validated FISH comparator test for the study. Concordances between the Oncomine Dx Target Test and the FISH test as well as clinical outcomes were reported in the Summary of Safety and Effectiveness Data. A total of 157 specimens were included. The PPA for Oncomine vs FISH was 80% (95% CI, 59 to 93) and NPA was 100% (95% CI, 97 to 100). For all ROS1-positive patients, as originally detected for enrollment into the ROS1 cohort, the response rate was 72% (95% CI, 58% to 84%). For ROS1-positive patients as detected by Oncomine, the response rate was 83% (95% CI, 36% to 99.6%).
 
 

CPT/HCPCS:
0022UTargeted genomic sequence analysis panel, non small cell lung neoplasia, DNA and RNA analysis, 23 genes, interrogation for sequence variants and rearrangements, reported as presence/absence of variants and associated therapy(ies) to consider
0239UTargeted genomic sequence analysis panel, solid organ neoplasm, cell free DNA, analysis of 311 or more genes, interrogation for sequence variants, including substitutions, insertions, deletions, select rearrangements, and copy number variations
0242UTargeted genomic sequence analysis panel, solid organ neoplasm, cell-free circulating DNA analysis of 55-74 genes, interrogation for sequence variants, gene copy number amplifications, and gene rearrangements
0388UOncology (non-small cell lung cancer), next generation dwquencing wtih identification of single nucleotide variants, copy number variants, insertions and deletions, and structural variants in 37 cancer-related genes, plasma, with report for alteration detection
0414UOncology (lung), augmentative algorithmic analysis of digitized whole slide imaging for 8 genes (ALK, BRAF, EGFR, ERBB2, MET, NTRK1-3, RET, ROS1), and KRAS G12C and PD-L1,if performed, formalin-fixed paraffin embedded (FFPE) tissue, reported as positive or negative for each biomarker
81191NTRK1 (neurotrophic receptor tyrosine kinase 1) (eg, solid tumors) translocation analysis
81192NTRK2 (neurotrophic receptor tyrosine kinase 2) (eg, solid tumors) translocation analysis
81193NTRK3 (neurotrophic receptor tyrosine kinase 3) (eg, solid tumors) translocation analysis
81194NTRK (neurotrophic tropomyosin receptor tyrosine kinase 1, 2, and 3) (eg, solid tumors) translocation analysis
81210BRAF (B Raf proto oncogene, serine/threonine kinase) (eg, colon cancer, melanoma), gene analysis, V600 variant(s)
81235EGFR (epidermal growth factor receptor) (eg, non small cell lung cancer) gene analysis, common variants (eg, exon 19 LREA deletion, L858R, T790M, G719A, G719S, L861Q)
81275KRAS (Kirsten rat sarcoma viral oncogene homolog) (eg, carcinoma) gene analysis; variants in exon 2 (eg, codons 12 and 13)
81276KRAS (Kirsten rat sarcoma viral oncogene homolog) (eg, carcinoma) gene analysis; additional variant(s) (eg, codon 61, codon 146)
81404Molecular pathology procedure, Level 5 (eg, analysis of 2-5 exons by DNA sequence analysis, mutation scanning or duplication/deletion variants of 6-10 exons, or characterization of a dynamic mutation disorder/triplet repeat by Southern blot analysis) ACADS (acyl-CoA dehydrogenase, C-2 to C-3 short chain) (eg, short chain acyl-CoA dehydrogenase deficiency), targeted sequence analysis (eg, exons 5 and 6) AQP2 (aquaporin 2 [collecting duct]) (eg, nephrogenic diabetes insipidus), full gene sequence ARX (aristaless related homeobox) (eg, X-linked lissencephaly with ambiguous genitalia, X-linked mental retardation), full gene sequence AVPR2 (arginine vasopressin receptor 2) (eg, nephrogenic diabetes insipidus), full gene sequence BBS10 (Bardet-Biedl syndrome 10) (eg, Bardet-Biedl syndrome), full gene sequence BTD (biotinidase) (eg, biotinidase deficiency), full gene sequence C10orf2 (chromosome 10 open reading frame 2) (eg, mitochondrial DNA depletion syndrome), full gene sequence CAV3 (caveolin 3) (eg, CAV3-related distal myopathy, limb-girdle muscular dystrophy type 1C), full gene sequence CD40LG (CD40 ligand) (eg, X-linked hyper IgM syndrome), full gene sequence CDKN2A (cyclin-dependent kinase inhibitor 2A) (eg, CDKN2A-related cutaneous malignant melanoma, familial atypical mole-malignant melanoma syndrome), full gene sequence CLRN1 (clarin 1) (eg, Usher syndrome, type 3), full gene sequence COX6B1 (cytochrome c oxidase subunit VIb polypeptide 1) (eg, mitochondrial respiratory chain complex IV deficiency), full gene sequence CPT2 (carnitine palmitoyltransferase 2) (eg, carnitine palmitoyltransferase II deficiency), full gene sequence CRX (cone-rod homeobox) (eg, cone-rod dystrophy 2, Leber congenital amaurosis), full gene sequence CYP1B1 (cytochrome P450, family 1, subfamily B, polypeptide 1) (eg, primary congenital glaucoma), full gene sequence EGR2 (early growth response 2) (eg, Charcot-Marie-Tooth), full gene sequence EMD (emerin) (eg, Emery-Dreifuss muscular dystrophy), duplication/deletion analysis EPM2A (epilepsy, progressive myoclonus type 2A, Lafora disease [laforin]) (eg, progressive myoclonus epilepsy), full gene sequence FGF23 (fibroblast growth factor 23) (eg, hypophosphatemic rickets), full gene sequence FGFR2 (fibroblast growth factor receptor 2) (eg, craniosynostosis, Apert syndrome, Crouzon syndrome), targeted sequence analysis (eg, exons 8, 10) FGFR3 (fibroblast growth factor receptor 3) (eg, achondroplasia, hypochondroplasia), targeted sequence analysis (eg, exons 8, 11, 12, 13) FHL1 (four and a half LIM domains 1) (eg, Emery-Dreifuss muscular dystrophy), full gene sequence FKRP (fukutin related protein) (eg, congenital muscular dystrophy type 1C [MDC1C], limb-girdle muscular dystrophy [LGMD] type 2I), full gene sequence FOXG1 (forkhead box G1) (eg, Rett syndrome), full gene sequence FSHMD1A (facioscapulohumeral muscular dystrophy 1A) (eg, facioscapulohumeral muscular dystrophy), evaluation to detect abnormal (eg, deleted) alleles FSHMD1A (facioscapulohumeral muscular dystrophy 1A) (eg, facioscapulohumeral muscular dystrophy), characterization of haplotype(s) (ie, chromosome 4A and 4B haplotypes) GH1 (growth hormone 1) (eg, growth hormone deficiency), full gene sequence GP1BB (glycoprotein Ib [platelet], beta polypeptide) (eg, Bernard-Soulier syndrome type B), full gene sequence (For common deletion variants of alpha globin 1 and alpha globin 2 genes, use 81257) HNF1B (HNF1 homeobox B) (eg, maturity-onset diabetes of the young [MODY]), duplication/deletion analysis HRAS (v-Ha-ras Harvey rat sarcoma viral oncogene homolog) (eg, Costello syndrome), full gene sequence HSD3B2 (hydroxy-delta-5-steroid dehydrogenase, 3 beta- and steroid delta-isomerase 2) (eg, 3-beta-hydroxysteroid dehydrogenase type II deficiency), full gene sequence HSD11B2 (hydroxysteroid [11-beta] dehydrogenase 2) (eg, mineralocorticoid excess syndrome), full gene sequence HSPB1 (heat shock 27kDa protein 1) (eg, Charcot-Marie-Tooth disease), full gene sequence INS (insulin) (eg, diabetes mellitus), full gene sequence KCNJ1 (potassium inwardly-rectifying channel, subfamily J, member 1) (eg, Bartter syndrome), full gene sequence KCNJ10 (potassium inwardly-rectifying channel, subfamily J, member 10) (eg, SeSAME syndrome, EAST syndrome, sensorineural hearing loss), full gene sequence LITAF (lipopolysaccharide-induced TNF factor) (eg, Charcot-Marie-Tooth), full gene sequence MEFV (Mediterranean fever) (eg, familial Mediterranean fever), full gene sequence MEN1 (multiple endocrine neoplasia I) (eg, multiple endocrine neoplasia type 1, Wermer syndrome), duplication/deletion analysis MMACHC (methylmalonic aciduria [cobalamin deficiency] cblC type, with homocystinuria) (eg, methylmalonic acidemia and homocystinuria), full gene sequence MPV17 (MpV17 mitochondrial inner membrane protein) (eg, mitochondrial DNA depletion syndrome), duplication/deletion analysis NDP (Norrie disease [pseudoglioma]) (eg, Norrie disease), full gene sequence NDUFA1 (NADH dehydrogenase [ubiquinone] 1 alpha subcomplex, 1, 7.5kDa) (eg, Leigh syndrome, mitochondrial complex I deficiency), full gene sequence NDUFAF2 (NADH dehydrogenase [ubiquinone] 1 alpha subcomplex, assembly factor 2) (eg, Leigh syndrome, mitochondrial complex I deficiency), full gene sequence NDUFS4 (NADH dehydrogenase [ubiquinone] Fe-S protein 4, 18kDa [NADH-coenzyme Q reductase]) (eg, Leigh syndrome, mitochondrial complex I deficiency), full gene sequence NIPA1 (non-imprinted in Prader-Willi/Angelman syndrome 1) (eg, spastic paraplegia), full gene sequence NLGN4X (neuroligin 4, X-linked) (eg, autism spectrum disorders), duplication/deletion analysis NPC2 (Niemann-Pick disease, type C2 [epididymal secretory protein E1]) (eg, Niemann-Pick disease type C2), full gene sequence NR0B1 (nuclear receptor subfamily 0, group B, member 1) (eg, congenital adrenal hypoplasia), full gene sequence PDX1 (pancreatic and duodenal homeobox 1) (eg, maturity-onset diabetes of the young [MODY]), full gene sequence PHOX2B (paired-like homeobox 2b) (eg, congenital central hypoventilation syndrome), full gene sequence PLP1 (proteolipid protein 1) (eg, Pelizaeus-Merzbacher disease, spastic paraplegia), duplication/deletion analysis PQBP1 (polyglutamine binding protein 1) (eg, Renpenning syndrome), duplication/deletion analysis PRNP (prion protein) (eg, genetic prion disease), full gene sequence PROP1 (PROP paired-like homeobox 1) (eg, combined pituitary hormone deficiency), full gene sequence PRPH2 (peripherin 2 [retinal degeneration, slow]) (eg, retinitis pigmentosa), full gene sequence PRSS1 (protease, serine, 1 [trypsin 1]) (eg, hereditary pancreatitis), full gene sequence RAF1 (v-raf-1 murine leukemia viral oncogene homolog 1) (eg, LEOPARD syndrome), targeted sequence analysis (eg, exons 7, 12, 14, 17) RET (ret proto-oncogene) (eg, multiple endocrine neoplasia, type 2B and familial medullary thyroid carcinoma), common variants (eg, M918T, 2647_2648delinsTT, A883F) RHO (rhodopsin) (eg, retinitis pigmentosa), full gene sequence RP1 (retinitis pigmentosa 1) (eg, retinitis pigmentosa), full gene sequence SCN1B (sodium channel, voltage-gated, type I, beta) (eg, Brugada syndrome), full gene sequence SCO2 (SCO cytochrome oxidase deficient homolog 2 [SCO1L]) (eg, mitochondrial respiratory chain complex IV deficiency), full gene sequence SDHC (succinate dehydrogenase complex, subunit C, integral membrane protein, 15kDa) (eg, hereditary paraganglioma-pheochromocytoma syndrome), duplication/deletion analysis SDHD (succinate dehydrogenase complex, subunit D, integral membrane protein) (eg, hereditary paraganglioma), full gene sequence SGCG (sarcoglycan, gamma [35kDa dystrophin-associated glycoprotein]) (eg, limb-girdle muscular dystrophy), duplication/deletion analysis SH2D1A (SH2 domain containing 1A) (eg, X-linked lymphoproliferative syndrome), full gene sequence SLC16A2 (solute carrier family 16, member 2 [thyroid hormone transporter]) (eg, specific thyroid hormone
81405Molecular pathology procedure, Level 6 (eg, analysis of 6-10 exons by DNA sequence analysis, mutation scanning or duplication/deletion variants of 11-25 exons, regionally targeted cytogenomic array analysis) Cytogenomic constitutional targeted microarray analysis of chromosome 22q13 by interrogation of genomic regions for copy number and single nucleotide polymorphism (SNP) variants for chromosomal abnormalities (When performing cytogenomic [genome-wide] analysis, for constitutional chromosomal abnormalities. See 81228, 81229, 81349)
81406Molecular pathology procedure, Level 7 (eg, analysis of 11-25 exons by DNA sequence analysis, mutation scanning or duplication/deletion variants of 26-50 exons) ACADVL (acyl-CoA dehydrogenase, very long chain) (eg, very long chain acyl-coenzyme A dehydrogenase deficiency), full gene sequence ACTN4 (actinin, alpha 4) (eg, focal segmental glomerulosclerosis), full gene sequence AFG3L2 (AFG3 ATPase family gene 3-like 2 [S. cerevisiae]) (eg, spinocerebellar ataxia), full gene sequence AIRE (autoimmune regulator) (eg, autoimmune polyendocrinopathy syndrome type 1), full gene sequence ALDH7A1 (aldehyde dehydrogenase 7 family, member A1) (eg, pyridoxine-dependent epilepsy), full gene sequence ANO5 (anoctamin 5) (eg, limb-girdle muscular dystrophy), full gene sequence ANOS1 (anosmin-1) (eg, Kallmann syndrome 1), full gene sequence APP (amyloid beta [A4] precursor protein) (eg, Alzheimer disease), full gene sequence ASS1 (argininosuccinate synthase 1) (eg, citrullinemia type I), full gene sequence ATL1 (atlastin GTPase 1) (eg, spastic paraplegia), full gene sequence ATP1A2 (ATPase, Na+/K+ transporting, alpha 2 polypeptide) (eg, familial hemiplegic migraine), full gene sequence ATP7B (ATPase, Cu++ transporting, beta polypeptide) (eg, Wilson disease), full gene sequence BBS1 (Bardet-Biedl syndrome 1) (eg, Bardet-Biedl syndrome), full gene sequence BBS2 (Bardet-Biedl syndrome 2) (eg, Bardet-Biedl syndrome), full gene sequence BCKDHB (branched-chain keto acid dehydrogenase E1, beta polypeptide) (eg, maple syrup urine disease, type 1B), full gene sequence BEST1 (bestrophin 1) (eg, vitelliform macular dystrophy), full gene sequence BMPR2 (bone morphogenetic protein receptor, type II [serine/threonine kinase]) (eg, heritable pulmonary arterial hypertension), full gene sequence BRAF (B-Raf proto-oncogene, serine/threonine kinase) (eg, Noonan syndrome), full gene sequence BSCL2 (Berardinelli-Seip congenital lipodystrophy 2 [seipin]) (eg, Berardinelli-Seip congenital lipodystrophy), full gene sequence BTK (Bruton agammaglobulinemia tyrosine kinase) (eg, X-linked agammaglobulinemia), full gene sequence CACNB2 (calcium channel, voltage-dependent, beta 2 subunit) (eg, Brugada syndrome), full gene sequence CAPN3 (calpain 3) (eg, limb-girdle muscular dystrophy [LGMD] type 2A, calpainopathy), full gene sequence CBS (cystathionine-beta-synthase) (eg, homocystinuria, cystathionine beta-synthase deficiency), full gene sequence CDH1 (cadherin 1, type 1, E-cadherin [epithelial]) (eg, hereditary diffuse gastric cancer), full gene sequence CDKL5 (cyclin-dependent kinase-like 5) (eg, early infantile epileptic encephalopathy), full gene sequence CLCN1 (chloride channel 1, skeletal muscle) (eg, myotonia congenita), full gene sequence CLCNKB (chloride channel, voltage-sensitive Kb) (eg, Bartter syndrome 3 and 4b), full gene sequence CNTNAP2 (contactin-associated protein-like 2) (eg, Pitt-Hopkins-like syndrome 1), full gene sequence COL6A2 (collagen, type VI, alpha 2) (eg, collagen type VI-related disorders), duplication/deletion analysis CPT1A (carnitine palmitoyltransferase 1A [liver]) (eg, carnitine palmitoyltransferase 1A [CPT1A] deficiency), full gene sequence CRB1 (crumbs homolog 1 [Drosophila]) (eg, Leber congenital amaurosis), full gene sequence CREBBP (CREB binding protein) (eg, Rubinstein-Taybi syndrome), duplication/deletion analysis DBT (dihydrolipoamide branched chain transacylase E2) (eg, maple syrup urine disease, type 2), full gene sequence DLAT (dihydrolipoamide S-acetyltransferase) (eg, pyruvate dehydrogenase E2 deficiency), full gene sequence DLD (dihydrolipoamide dehydrogenase) (eg, maple syrup urine disease, type III), full gene sequence DSC2 (desmocollin) (eg, arrhythmogenic right ventricular dysplasia/cardiomyopathy 11), full gene sequence DSG2 (desmoglein 2) (eg, arrhythmogenic right ventricular dysplasia/cardiomyopathy 10), full gene sequence DSP (desmoplakin) (eg, arrhythmogenic right ventricular dysplasia/cardiomyopathy 8), full gene sequence EFHC1 (EF-hand domain [C-terminal] containing 1) (eg, juvenile myoclonic epilepsy), full gene sequence EIF2B3 (eukaryotic translation initiation factor 2B, subunit 3 gamma, 58kDa) (eg, leukoencephalopathy with vanishing white matter), full gene sequence EIF2B4 (eukaryotic translation initiation factor 2B, subunit 4 delta, 67kDa) (eg, leukoencephalopathy with vanishing white matter), full gene sequence EIF2B5 (eukaryotic translation initiation factor 2B, subunit 5 epsilon, 82kDa) (eg, childhood ataxia with central nervous system hypomyelination/vanishing white matter), full gene sequence ENG (endoglin) (eg, hereditary hemorrhagic telangiectasia, type 1), full gene sequence EYA1 (eyes absent homolog 1 [Drosophila]) (eg, branchio-oto-renal [BOR] spectrum disorders), full gene sequence F8 (coagulation factor VIII) (eg, hemophilia A), duplication/deletion analysis FAH (fumarylacetoacetate hydrolase [fumarylacetoacetase]) (eg, tyrosinemia, type 1), full gene sequence FASTKD2 (FAST kinase domains 2) (eg, mitochondrial respiratory chain complex IV deficiency), full gene sequence FIG4 (FIG4 homolog, SAC1 lipid phosphatase domain containing [S. cerevisiae]) (eg, Charcot-Marie-Tooth disease), full gene sequence FTSJ1 (FtsJ RNA methyltransferase homolog 1 [E. coli]) (eg, X-linked mental retardation 9), full gene sequence FUS (fused in sarcoma) (eg, amyotrophic lateral sclerosis), full gene sequence GAA (glucosidase, alpha; acid) (eg, glycogen storage disease type II [Pompe disease]), full gene sequence GALC (galactosylceramidase) (eg, Krabbe disease), full gene sequence GALT (galactose-1-phosphate uridylyltransferase) (eg, galactosemia), full gene sequence GARS (glycyl-tRNA synthetase) (eg, Charcot-Marie-Tooth disease), full gene sequence GCDH (glutaryl-CoA dehydrogenase) (eg, glutaricacidemia type 1), full gene sequence GCK (glucokinase [hexokinase 4]) (eg, maturity-onset diabetes of the young [MODY]), full gene sequence GLUD1 (glutamate dehydrogenase 1) (eg, familial hyperinsulinism), full gene sequence GNE (glucosamine [UDP-N-acetyl]-2-epimerase/N-acetylmannosamine kinase) (eg, inclusion body myopathy 2 [IBM2], Nonaka myopathy), full gene sequence GRN (granulin) (eg, frontotemporal dementia), full gene sequence HADHA (hydroxyacyl-CoA dehydrogenase/3-ketoacyl-CoA thiolase/enoyl-CoA hydratase [trifunctional protein] alpha subunit) (eg, long chain acyl-coenzyme A dehydrogenase deficiency), full gene sequence HADHB (hydroxyacyl-CoA dehydrogenase/3-ketoacyl-CoA thiolase/enoyl-CoA hydratase [trifunctional protein], beta subunit) (eg, trifunctional protein deficiency), full gene sequence HEXA (hexosaminidase A, alpha polypeptide) (eg, Tay-Sachs disease), full gene sequence HLCS (HLCS holocarboxylase synthetase) (eg, holocarboxylase synthetase deficiency), full gene sequence HMBS (hydroxymethylbilane synthase) (eg, acute intermittent porphyria), full gene sequence HNF4A (hepatocyte nuclear factor 4, alpha) (eg, maturity-onset diabetes of the young [MODY]), full gene sequence IDUA (iduronidase, alpha-L-) (eg, mucopolysaccharidosis type I), full gene sequence INF2 (inverted formin, FH2 and WH2 domain containing) (eg, focal segmental glomerulosclerosis), full gene sequence IVD (isovaleryl-CoA dehydrogenase) (eg, isovaleric acidemia), full gene sequence JAG1 (jagged 1) (eg, Alagille syndrome), duplication/deletion analysis JUP (junction plakoglobin) (eg, arrhythmogenic right ventricular dysplasia/cardiomyopathy 11), full gene sequence KCNH2 (potassium voltage-gated channel, subfamily H [eag-related], member 2) (eg, short QT syndrome, long QT syndrome), full gene sequence KCNQ1 (potassium voltage-gated channel, KQT-like subfamily, member 1) (eg, short QT syndrome, long QT syndrome), full gene sequence KCNQ2 (potassium voltage-gated channel, KQT-like subfamily, member 2) (eg, epileptic encephalopathy), full gene sequence LDB3 (LIM domain binding 3) (eg, familial dilated cardiomyopathy, myofibrillar myopathy), full gene sequence LDLR (low den
81479Unlisted molecular pathology procedure
88365In situ hybridization (eg, FISH), per specimen; initial single probe stain procedure

References: Ahn MJ, Park BB, Ahn JS, et al.(2008) Are there any ethnic differences in molecular predictors of erlotinib efficacy in advanced non-small cell lung cancer? Clin Cancer Res. Jun 15 2008;14(12):3860-3866. PMID 18559606

Amann JM, Lee JW, Roder H, et al.(2010) Genetic and proteomic features associated with survival after treatment with erlotinib in first-line therapy of non-small cell lung cancer in Eastern Cooperative Oncology Group 3503. J Thorac Oncol. Feb 2010;5(2):169-178. PMID 20035238

Amgen, Inc(2021) LUMAKRAS™ (sotorasib) U.S. Food and Drug Administration website. https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/214665s000lbl.pdf. Revised May 2021. Accessed Aug 24, 2021.

An N, Zhang Y, Niu H, et al.(2016) EGFR-TKIs versus taxanes agents in therapy for nonsmall-cell lung cancer patients: A PRISMA-compliant systematic review with meta-analysis and meta-regression. Medicine (Baltimore). Dec 2016;95(50):e5601. PMID 27977598

Bergethon K, Shaw AT, Ou SH, et al.(2012) ROS1 rearrangements define a unique molecular class of lung cancers. J Clin Oncol. Mar 10 2012;30(8):863-870. PMID 22215748

Boehringer Ingelheim Pharmaceuticals, Inc.(2014) Gilotrif™ (afatinib) tablets for oral use prescribing information, November 2013. http://www.gilotrif.com/. Accessed July, 2014.

Boldrini L, Ali G, Gisfredi S, et al.(2009) Epidermal growth factor receptor and K-RAS mutations in 411 lung adenocarcinoma: a population-based prospective study. Oncol Rep. Oct 2009;22(4):683-691. PMID 19724844

Camidge DR, Bang YJ, Kwak EL, et al.(2012) Activity and safety of crizotinib in patients with ALK-positive non-smallcell lung cancer: updated results from a phase 1 study. Lancet Oncol. Oct 2012;13(10):1011-1019. PMID 22954507

Camidge DR, Kim DW, Tiseo M, et al.(2018) Exploratory analysis of brigatinib activity in patients with anaplastic lymphoma kinase-positive non-small-cell lung cancer and brain metastases in two clinical trials. J Clin Oncol. Sep 10 2018;36(26):2693-2701. PMID 29768119

Chen G, Feng J, Zhou C, et al.(2013) Quality of life (QoL) analyses from OPTIMAL (CTONG-0802), a phase III, randomised, open-label study of first-line erlotinib versus chemotherapy in patients with advanced EGFR mutation-positive non-small-cell lung cancer (NSCLC). Ann Oncol. June 1, 2013 2013;24(6):1615-1622. PMID

Cheng Y, Mok TS, Zhou X, et al.(2021) Safety and efficacy of first-line dacomitinib in Asian patients with EGFR mutation-positive non-small cell lung cancer: Results from a randomized, open-label, phase 3 trial (ARCHER 1050). Lung Cancer. Apr 2021; 154:176-185. PMID 33721611

Crequit P, Chaimani A, Yavchitz A, et al.(2017) Comparative efficacy and safety of second-line treatments for advanced non-small cell lung cancer with wild-type or unknown status for epidermal growth factor receptor: a systematic review and network meta-analysis. BMC Med. Oct 30 2017;15(1):193. PMID 29082855

Dobashi Y, Suzuki S, Kimura M, et al.(2011) Paradigm of kinase-driven pathway downstream of epidermal growth factor receptor/Akt in human lung carcinomas. Hum Pathol. Feb 2011;42(2):214-226. PMID 21040950

Doebele RC, Drilon A, Paz-Ares L, et al.(2020) Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: integrated analysis of three phase 1-2 trials. Lancet Oncol. Feb 2020; 21(2): 271-282. PMID 31838007

Drilon A, Laetsch TW, Kummar S, et al.(2018) Efficacy of Larotrectinib in TRK Fusion-Positive Cancers in Adults and Children. N Engl J Med. Feb 22 2018; 378(8): 731-739. PMID 29466156

Drilon A, Oxnard GR, Tan DSW, et al.(2020) Efficacy of Selpercatinib in RET Fusion-Positive Non-Small-Cell Lung Cancer. N Engl J Med. Aug 27 2020; 383(9): 813-824. PMID 32846060

Drilon A, Wang L, Hasanovic A, et al.(2013) Response to cabozantinib in patients with RET fusion-positive lung adenocarcinomas. Cancer Discov. Jun 2013;3(6):630-635. PMID 23533264

Drilon A, Wang L, Hasanovic A, et al.(2013) Response to Cabozantinib in patients with RET fusionpositive lung adenocarcinomas. Cancer Discov. Jun 2013;3(6):630-635. PMID 23533264

Dziadziuszko R, Hung T, Wang K, et al.(2022) Pre- and post-treatment blood-based genomic landscape of patients with ROS1 or NTRK fusion-positive solid tumours treated with entrectinib. Mol Oncol. May 2022; 16(10): 2000-2014. PMID 35338679

Dziadziuszko R, Mok T, Peters S, et al.(2021) Blood First Assay Screening Trial (BFAST) in Treatment-Naive Advanced or Metastatic NSCLC: Initial Results of the Phase 2 ALK-Positive Cohort. J Thorac Oncol. Jul 24 2021. PMID 34311110

Eberhard DA, Johnson BE, Amler LC, et al.(2005) Mutations in the epidermal growth factor receptor and in KRAS are predictive and prognostic indicators in patients with non-small-cell lung cancer treated with chemotherapy alone and in combination with erlotinib. J Clin Oncol. 2005;23(25):5900-5909. PMID

Eberhard DA, Johnson BE, Amler LC, et al.(2005) Mutations in the epidermal growth factor receptor and in KRAS are predictive and prognostic indicators in patients with non-small-cell lung cancer treated with chemotherapy alone and in combination with erlotinib. J Clin Oncol. Sep 1 2005;23(25):5900-5909. PMID 16043828

Eli Lilly and Company.(2020) Highlights of Prescribing Information: CYRAMZA (ramucirumab) injection, for intravenous use. 2020; https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/125477s034lbl.pdf. Accessed October 3, 2022.

Fang W, Zhang J, Liang W, et al.(2013) Efficacy of epidermal growth factor receptor-tyrosine kinase inhibitors for Chinese patients with squamous cell carcinoma of lung harboring EGFR mutation. J Thorac Dis. 2013;5(5):585-592. PMID

Fathi AT, Brahmer JR(2008) for advanced stage non-small cell lung cancer. Semin Thorac Cardiovasc Surg. Fall 2008;20(3):210-216. PMID 19038730

Felip E, Rojo F, Reck M, et al.(2008) A phase II pharmacodynamic study of erlotinib in patients with advanced non-small cell lung cancer previously treated with platinum-based chemotherapy. Clin Cancer Res. Jun 15 2008;14(12):3867-3874. PMID 18559607

Fiala O, Pesek M, Finek J, et al.(2013) Gene mutations in squamous cell NSCLC: insignificance of EGFR, KRAS and PIK3CA mutations in prediction of EGFR-TKI treatment efficacy. Anticancer Res. Apr 2013;33(4):1705-1711. PMID 23564819

Food and Drug Adinistration (FDA).(2016) Summary of Safety and Effectiveness Data (SSED):Oncomine™ DX Target Test. 2017; https://www.accessdata.fda.gov/cdrh_docs/pdf16/p160045b.pdf. Accessed September 1, 2017.

Food and Drug Administration (FDA).(2011) Summary of Safety and Effectiveness Data (SSED): Vysis ALK Break Apart FISH Probe Kit. 2011; https://www.accessdata.fda.gov/cdrh_docs/pdf11/P110012b.pdf. Accessed September 6, 2017.

Food and Drug Administration (FDA).(2013) Summary of Safety and Effectiveness Data (SSED): cobas® EGFR Mutation Test. 2013; https://www.accessdata.fda.gov/cdrh_docs/pdf12/P120019b.pdf. Accessed September 5, 2017.

Food and Drug Administration (FDA).(2013) Summary of Safety and Effectiveness Data (SSED): therascreen® EGFR RGQ PCR Kit. 2013; https://www.accessdata.fda.gov/cdrh_docs/pdf12/P120022b.pdf. Accessed September 5, 2017.

Food and Drug Administration (FDA).(2014) Highlights of Prescribing Information: Mekinist. 2017; https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/204114s001lbl.pdf. Accessed August 31, 2017.

Food and Drug Administration (FDA).(2015) Summary of Safety and Effectiveness Data (SSED): cobas® EGFR Mutation Test v2. 2015; https://www.accessdata.fda.gov/cdrh_docs/pdf12/P120019S007b.pdf. Accessed September 5, 2017.

Food and Drug Administration (FDA).(2015) Summary of Safety and Effectiveness Data (SSED): Ventana ALK (D5F3) CDx Assay. 2015; https://www.accessdata.fda.gov/cdrh_docs/pdf14/P140025B.pdf. Accessed September 6, 2017.

Food and Drug Administration (FDA).(2017) Highlights of Prescribing Information: Tafinlar. 2017; https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/202806s002lbl.pdf. Accessed August 31, 2017.

Food and Drug Administration (FDA).(2018) Multi-Disciplinary Review and Evaluation: VIZIMPRO / dacomitinib [NDA 211288]. 2018; https://www.accessdata.fda.gov/drugsatfda_docs/nda/2018/211288Orig1s000MultidisciplineR.pdf. Accessed October 1, 2022.

Food and Drug Administration (FDA).(2020) Summary of Safety and Effectiveness Data (SSED) FoundationOne Liquid CDx. 2020; https://www.accessdata.fda.gov/cdrh_docs/pdf19/P190032B.pdf. Accessed September 26, 2022.

Food and Drug Administration (FDA).(2020) Summary of Safety and Effectiveness Data. FoundationOne Liquid CDx. 2020. https://www.accessdata.fda.gov/cdrh_docs/pdf20/P200006B.pdf. Accessed October 29, 2021.

Food and Drug Administration (FDA).(2021) FDA approves cemiplimab-rwlc for non-small cell lung cancer with high PD-L1expression. February 22, 2021. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-cemiplimab-rwlc-non-small-cell-lung-cancer-high-pd-l1-expression. Accessed October 7, 2022.

Food and Drug Administration (FDA).(2021) Multi-disciplinary Review and Evaluation: Mobocertinib (formerly TAK-788, AP32788) [NDA 215310]. 2021; https://www.accessdata.fda.gov/drugsatfda_docs/nda/2021/215310Orig1s000MultidisciplineR.pdf.Accessed October 2, 2022.

Food and Drug Administration (FDA).(2021) Multi-disciplinary Review and Evaluation: RYBREVANT (amivantamab-vmjw) [BLA761210]. 2021; https://www.accessdata.fda.gov/drugsatfda_docs/nda/2021/761210Orig1s000MultidisciplineR.pdf. Accessed on October 4, 2022.

Food and Drug Administration (FDA).(2021) Summary of Safety and Effectiveness Data (SSED): Guardant360 CDx [P200010/S002]. 2021; https://www.accessdata.fda.gov/cdrh_docs/pdf20/P200010S002B.pdf. Accessed October 9, 2022.

Food and Drug Administration (FDA).(2022) NDA/BLA Multi-disciplinary Review and Evaluation [BLA 761139] ENHERTU (fam-trastuzumab deruxtecan-nxki). 2019; https://www.accessdata.fda.gov/drugsatfda_docs/nda/2019/761139Orig1s000MultidisciplineR.pdf. Accessed October 6, 2022.

Forbes SA, Bhamra G, Bamford S, et al.(2008) The Catalogue of Somatic Mutations in Cancer (COSMIC). Curr Protoc Hum Genet. Apr 2008;Chapter 10:Unit 10 11. PMID 18428421

Frampton GM, Fichtenholtz A, Otto GA, et al.(2013) Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat Biotechnol. Nov 2013;31(11):1023-1031. PMID 24142049

Garassino MC, Martelli O, Broggini M, et al.(2013) Erlotinib versus docetaxel as second-line treatment of patients with advanced non-small-cell lung cancer and wild-type EGFR tumours (TAILOR): a randomised controlled trial. Lancet Oncol. 2013;14(10):981-988. PMID

Gautschi O, Pauli C, Strobel K, et al.(2012) A patient with BRAF V600E lung adenocarcinoma responding to vemurafenib. J Thorac Oncol. Oct 2012;7(10):e23-24. PMID 22743296

Gautschi O, Pauli C, Strobel K, et al.(2012) A patient with BRAF V600E lung adenocarcinoma responding to vemurafenib. J Thorac Oncol. Oct 2012;7(10):e23-24. PMID 22743296

Gettinger SN, Bazhenova LA, Langer CJ, et al.(2016) Activity and safety of brigatinib in ALK-rearranged non-small-cell lung cancer and other malignancies: a single-arm, open-label, phase 1/2 trial. Lancet Oncol. Dec 2016;17(12):1683-1696. PMID 27836716

Giaccone G, Gallegos RM, Le CT, et al.(2006) Erlotinib for frontline treatment of advanced non-small cell lung cancer: a phase II study. Clin Cancer Res. 2006;12(20):6049-6055. PMID

Giaccone G, Gallegos Ruiz M, Le Chevalier T, et al.(2006) Erlotinib for frontline treatment of advanced non-small cell lung cancer: a phase II study. Clin Cancer Res. Oct 15 2006;12(20 Pt 1):6049- 6055. PMID 17062680

Guan JL, Zhong WZ, An SJ, et al.(2013) KRAS mutation in patients with lung cancer: a predictor for poor prognosis but not for EGFR-TKIs or chemotherapy. Ann Surg Oncol. Apr 2013;20(4):1381-1388. PMID 23208128

Han B, Jin B, Chu T, et al.(2017) Combination of chemotherapy and gefitinib as first-line treatment for patients with advanced lung adenocarcinoma and sensitive EGFR mutations: A randomized controlled trial. Int J Cancer. Sep 15 2017;141(6):1249-1256. PMID 28560853

Hanna N, Johnson D, Temin S, et al.(2017) Systemic therapy for stage IV Non-small-cell lung cancer: American Society of Clinical Oncology Clinical Practice Guideline Update. J Clin Oncol. Aug 14 2017:JCO2017746065. PMID 28806116

Hanna NH, Robinson AG, Temin S, et al.(2021) Therapy for Stage IV Non-Small-Cell Lung Cancer With Driver Alterations: ASCO and OH(CCO) Joint Guideline Update. J Clin Oncol. Mar 20 2021; 39(9): 1040-1091. PMID 33591844

Hellmann MD, Ciuleanu TE, Pluzanski A, et al.(2018) Nivolumab plus Ipilimumab in Lung Cancer with a High Tumor Mutational Burden. N Engl J Med. May 31 2018; 378(22): 2093-2104. PMID 29658845

Hellmann MD, Ciuleanu TE, Pluzanski A, et al.(2018) Nivolumab plus Ipilimumab in Lung Cancer with a High Tumor Mutational Burden. N Engl J Med. May 31 2018; 378(22): 2093-2104. PMID 29658845

Hellmann MD, Paz-Ares L, Bernabe Caro R, et al.(2019) Nivolumab plus Ipilimumab in Advanced Non-Small-Cell Lung Cancer. N Engl J Med. Nov 21 2019; 381(21): 2020-2031. PMID 31562796

Herbst RS, Giaccone G, de Marinis F, et al.(2020) Atezolizumab for First-Line Treatment of PD-L1-Selected Patients with NSCLC. N Engl J Med. Oct 01 2020; 383(14): 1328-1339. PMID 32997907

Herbst RS, Prager D, Hermann R, et al.(2005) TRIBUTE: a phase III trial of erlotinib hydrochloride (OSI- 774) combined with carboplatin and paclitaxel chemotherapy in advanced non-small-cell lung cancer. J Clin Oncol. 2005;23(25):5856-5858. PMID

Hida T, Nokihara H, Kondo M, et al.(2017) Alectinib versus crizotinib in patients with ALK-positive non-small-cell lung cancer (J-ALEX): an open-label, randomised phase 3 trial. Lancet. Jul 01 2017;390(10089):29-39. PMID 28501140

Hong DS, DuBois SG, Kummar S, et al.(2020) Larotrectinib in patients with TRK fusion-positive solid tumours: a pooled analysis of three phase 1/2 clinical trials. Lancet Oncol. Apr 2020; 21(4): 531-540. PMID 32105622

Husain H, Pavlick DC, Fendler BJ, et al.(2022) Tumor Fraction Correlates With Detection of Actionable Variants Across 23,000 Circulating Tumor DNA Samples. JCO Precis Oncol. Oct 2022; 6: e2200261. PMID 36265119

Hyman DM, Puzanov I, Subbiah V, et al.(2015) Vemurafenib in multiple nonmelanoma cancers with BRAF V600 mutations. N Engl J Med. Aug 20 2015;373(8):726-736. PMID 26287849

Inoue A, Kobayashi K, Maemondo M, et al.(2013) Updated overall survival results from a randomized phase III trial comparing gefitinib with carboplatin-paclitaxel for chemo-naive non-small cell lung cancer with sensitive EGFR gene mutations (NEJ002). Ann Oncol. Jan 2013;24(1):54-59. PMID 22967997

Jackman DM, Miller VA, Cioffredi LA, et al.(2009) Impact of epidermal growth factor receptor and KRAS mutations on clinical outcomes in previously untreated non-small cell lung cancer patients: results of an online tumor registry of clinical trials. Clin Cancer Res. Aug 15 2009;15(16):5267-5273. PMID 19671843

Jackman DM, Yeap BY, Lindeman NI, et al.(2007) Phase II clinical trial of chemotherapy-naive patients > or = 70 years of age treated with erlotinib for advanced non-small-cell lung cancer. J Clin Oncol. 2007;25(7):760-766. PMID

Jackman DM, Yeap BY, Lindeman NI, et al.(2007) Phase II clinical trial of chemotherapy-naive patients > or = 70 years of age treated with erlotinib for advanced non-small-cell lung cancer. J Clin Oncol. Mar 1 2007;25(7):760-766. PMID 17228019

Jassem J, Dziadziuszko R.(2013) EGFR inhibitors for wild-type EGFR NSCLC: to use or not to use? Lancet Oncol. 2013;14(10):916-917. PMID

Jazieh AR, Al Sudairy R, Abu-Shraie N, et al.(2013) Erlotinib in wild type epidermal growth factor receptor non-small cell lung cancer: A systematic review. Ann Thorac Med. Oct 2013;8(4):204-208. PMID 24250733

Kalemkerian GP, Narula N, Kennedy EB.(2018) Molecular testing guideline for the selection of lung cancer patients for treatment with targeted tyrosine kinase inhibitors: American Society of Clinical Oncology Endorsement Summary of the College of American Pathologists/International Association for the Study of Lung Cancer/Association for Molecular Pathology Clinical Practice Guideline Update. J Oncol Pract. May 2018;14(5):323-327. PMID 29589987

Katakami N, Atagi S, Goto K, et al(2013) LUX-Lung 4: A Phase II Trial of Afatinib in Patients With Advanced Non–Small-Cell Lung Cancer Who Progressed During Prior Treatment With Erlotinib, Gefitinib, or Both. J Clin Oncol. September 20, 2013 2013;31(27):3335-3341. PMID

Keedy VL, Temin S, Somerfield MR, et al.(2011) American Society of Clinical Oncology provisional clinical opinion: epidermal growth factor receptor (EGFR) Mutation testing for patients with advanced non-small-cell lung cancer considering first-line EGFR tyrosine kinase inhibitor therapy. J Clin Oncol. May 20 2011;29(15):2121-2127. PMID 21482992

Khambata-Ford S, Harbison CT, Hart LL, et al.(2010) Analysis of potential predictive markers of cetuximab benefit in BMS099, a phase III study of cetuximab and first-line taxane/carboplatin in advanced non-small-cell lung cancer. J Clin Oncol. 2010;28(6):918-927. PMID

Kim DW, Tiseo M, Ahn MJ, et al.(2017) Brigatinib in patients with crizotinib-refractory anaplastic lymphoma kinasepositive non-small-cell lung cancer: a randomized, multicenter phase II trial. J Clin Oncol. Aug 01 2017;35(22):2490-2498. PMID 28475456

Kim HR, Lim SM, Kim HJ, et al.(2013) The frequency and impact of ROS1 rearrangement on clinical outcomes in never smokers with lung adenocarcinoma. Ann Oncol. Sep 2013;24(9):2364-2370. PMID 23788756

Kim HR, Lim SM, Kim HJ, et al.(2013) The frequency and impact of ROS1 rearrangement on clinical outcomes in never smokers with lung adenocarcinoma. Ann Oncol. Sep 2013;24(9):2364-2370. PMID 23788756

Kwak EL, Bang YJ, Camidge DR, et al.(2010) Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N Engl J Med. Oct 28 2010;363(18):1693-1703. PMID 20979469

Lee CK, Brown C, Gralla RJ, et al.(2013) Impact of EGFR inhibitor in non-small cell lung cancer on progression-free and overall survival: a meta-analysis. J Natl Cancer Inst. May 1 2013;105(9):595-605. PMID 3594426

Lim SM, Kim HR, Lee JS, et al.(2017) Open-label, multicenter, phase ii study of ceritinib in patients with non-small-cell lung cancer harboring ROS1 rearrangement. J Clin Oncol. Aug 10 2017;35(23):2613-2618. PMID 28520527

Lin JZ, Ma SK, Wu SX, et al.(2018) A network meta-analysis of nonsmall-cell lung cancer patients with an activating EGFR mutation: Should osimertinib be the first-line treatment? Medicine (Baltimore). Jul 2018;97(30):e11569. PMID 30045282

Linardou H, Dahabreh IJ, Kanaloupiti D, et al.(2008) Assessment of somatic k-RAS mutations as a mechanism associated with resistance to EGFR-targeted agents: A systematic review and metaanalysis of studies in advanced non-small-cell lung cancer and metastatic colorectal cancer. Lancet Oncol. 2008;9(10):962-972. PMID

Lindeman NI, Cagle PT, Aisner DL, et al.(2018) Updated Molecular Testing Guideline for the Selection of Lung Cancer Patients for Treatment With Targeted Tyrosine Kinase Inhibitors: Guideline From the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology. J Mol Diagn. Mar 2018;20(2):129-159. PMID 29398453

Lindeman NI, Cagle PT, Beasley MB, et al.(2013) Molecular testing guideline for selection of lung cancer patients for EGFR and ALK tyrosine kinase inhibitors: guideline from the College of American Pathologists, International Association for the Study of Lung Cancer, and Association for Molecular Pathology. Arch Pathol Lab Med. Jun 2013;137(6):828-860. PMID 23551194

Lynch TJ, Bell DW, Sordella R, et al.(2004) Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med. May 20 2004;350(21):2129-2139. PMID 15118073

Lynch TJ, Patel T, Dreisbach L, et al.(2010) Cetuximab and first-line taxane/carboplatin chemotherapy in advanced non-small-cell lung cancer: Results of the randomized multicenter phase III trial BMS099. J Clin Oncol. 2010;28(6):911-917. PMID

Maemondo M, Inoue A, Kobayashi K, et al.(2010) Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Engl J Med. Jun 24 2010;362(25):2380-2388. PMID 20573926

Mao C, Qiu LX, Liao RY, et al.(2010) KRAS mutations and resistance to EGFR-TKIs treatment in patients with non-small cell lung cancer: a meta-analysis of 22 studies. Lung Cancer. 2010;69(3):272-278. PMID

Marabelle A, Fakih M, Lopez J, et al.(2020) Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol. Oct 2020; 21(10): 1353-1365. PMID 32919526

Martin P, Leighl NB, Tsao MS, et al.(2013) KRAS mutations as prognostic and predictive markers in non-small cell lung cancer. J Thorac Oncol. May 2013;8(5):530-542. PMID 23524403.

Martoni A, Marino A, Sperandi F, et al.(2005) Multicentre randomised phase III study comparing the same dose and schedule of cisplatin plus the same schedule of vinorelbine or gemcitabine in advanced non-small cell lung cancer. Eur J Cancer. Jan 2005;41(1):81-92. PMID 15617993

Mazieres J, Peters S, Lepage B, et al.(2013) Lung cancer that harbors an HER2 mutation: epidemiologic characteristics and therapeutic perspectives. J Clin Oncol. Jun 1 2013;31(16):1997-2003. PMID 23610105

Mazieres J, Zalcman G, Crino L, et al.(2015) Crizotinib therapy for advanced lung adenocarcinoma and a ROS1 rearrangement: results from the EUROS1 cohort. J Clin Oncol. Mar 20 2015;33(9):992-999. PMID 25667280

Miller VA, Hirsh V, Cadranel J, et al.(2012) Afatinib versus placebo for patients with advanced, metastatic non-small-cell lung cancer after failure of erlotinib, gefitinib, or both, and one or two lines of chemotherapy (LUX-Lung 1): a phase 2b/3 randomised trial. Lancet Oncol. 2012;13(5):528-538. PMID

Miller VA, Riely GJ, Zakowski MF, et al.(2008) Molecular characteristics of bronchioloalveolar carcinoma and adenocarcinoma, bronchioloalveolar carcinoma subtype, predict response to erlotinib. J Clin Oncol. 2008;26(9):1472-1478. PMID

Miller VA, Riely GJ, Zakowski MF, et al.(2008) Molecular characteristics of bronchioloalveolar carcinoma and adenocarcinoma, bronchioloalveolar carcinoma subtype, predict response to erlotinib. J Clin Oncol. Mar 20 2008;26(9):1472-1478. PMID 18349398

Mitsudomi T, Kosaka T, Yatabe Y.(2006) Biological and clinical implications of EGFR mutations in lung cancer. Int J Clin Oncol. Jun 2006;11(3):190-198. PMID 16850125

Mitsudomi T, Morita S, Yatabe Y, et al.(2010) Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor (WJTOG3405): an open label, randomised phase 3 trial. Lancet Oncol. Feb 2010;11(2):121-128. PMID 20022809

Mok TS, Wu YL, Ahn MJ, et al.(2017) Osimertinib or Platinum-Pemetrexed in EGFR T790M-Positive Lung Cancer. N Engl J Med. Feb 16 2017;376(7):629-640. PMID 27959700

Mok TS, Wu YL, Thongprasert S, et al.(2009) Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med. Sep 03 2009;361(10):947-957. PMID 19692680

Mujoomdar M, Moulton K, Spry C.(2010) Epidermal Growth Factor Receptor Mutation Analysis in Advanced Non-Small Cell Lung Cancer: A Review of the Clinical Effectiveness and Guidelines. Ottawa: Canadian Agency for Drugs and Technologies in Health; 2010. http://www.cadth.ca/en/search?q=epidermal+growth+factor+receptor. Accessed July, 2014.

Nakagawa K, Nadal E, Garon EB, et al.(2021) RELAY Subgroup Analyses by EGFR Ex19del and Ex21L858R Mutations for Ramucirumab Plus Erlotinib in Metastatic Non-Small Cell Lung Cancer. Clin Cancer Res. Oct 01 2021; 27(19): 5258-5271.PMID 34301751

National Comprehensive Cancer Network (NCCN).(2022) NCCN Clinical Practice Guidelines in Oncology: Non-Small Cell Lung Cancer. Version 5.2022. https://www.nccn.org/professionals/physician_gls/pdf/nscl.pdf. Accessed October 17, 2022.

National Comprehensive Cancer Network.(2014) NCCN Clinical Practice Guidelines in Oncology: Non- Small Cell Lung Cancer, version 2.2014 (discussion update in progress). http://www.nccn.org/professionals/physician_gls/PDF/nscl.pdf. Accessed July, 2014.

O'Byrne KJ, Gatzemeier U, Bondarenko I, et al.(2011) Molecular biomarkers in non-small-cell lung cancer: a retrospective analysis of data from the phase 3 FLEX study. Lancet Oncol. Aug 2011;12(8):795-805. PMID 21782507

Ou SH, Ahn JS, De Petris L, et al.(2016) Alectinib in Crizotinib-refractory ALK-rearranged non-small-cell lung cancer: a phase II global study. J Clin Oncol. Mar 01 2016;34(7):661-668. PMID 26598747

Owen DH, Singh N, Ismaila N, et al.(2023) Therapy for Stage IV Non-Small-Cell Lung Cancer With Driver Alterations: ASCO Living Guideline, Version 2023.2. J Clin Oncol. Aug 20 2023; 41(24): e63-e72. PMID 37433095

Paez JG, Janne PA, Lee JC, et al(2004) EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science. Jun 4 2004;304(5676):1497-1500. PMID 15118125

Paik PK, Varghese AM, Sima CS, et al.(2012) Response to erlotinib in patients with EGFR mutant advanced non-small cell lung cancers with a squamous or squamous-like component. Mol Cancer Ther. Nov 2012;11(11):2535-2540. PMID 22896669

Pao W, Wang TY, Riely GJ, et al.(2005) KRAS mutations and primary resistance of lung adenocarcinomas to gefitinib or erlotinib. PLoS Med. 2005;2(1):57-61. PMID005

Park K, Tan EH, O'Byrne K, et al.(2016) Afatinib versus gefitinib as first-line treatment of patients with EGFR mutation-positive non-small-cell lung cancer (LUX-Lung 7): a phase 2B, open-label, randomised controlled trial. Lancet Oncol. May 2016;17(5):577-589. PMID 27083334

Park SH, Ha SY, Lee JI, et al.(2009) Epidermal growth factor receptor mutations and the clinical outcome in male smokers with squamous cell carcinoma of lung. J Korean Med Sci. Jun 2009;24(3):448-452. PMID 19543508

Paz-Ares L, Soulieres D, Melezinek I, et al.(2010) Clinical outcomes in non-small-cell lung cancer patients with EGFR mutations: pooled analysis. J Cell Mol Med. Jan 2010;14(1-2):51-69. PMID 20015198

Peters S, Camidge DR, Shaw AT, et al.(2017) Alectinib versus Crizotinib in Untreated ALK-Positive Non-Small-Cell Lung Cancer. N Engl J Med. Aug 31 2017;377(9):829-838. PMID 28586279

Peters S, Michielin O, Zimmermann S.(2013) Dramatic response induced by vemurafenib in a BRAF V600E-mutated lung adenocarcinoma. J Clin Oncol. Jul 10 2013;31(20):e341-344. PMID 23733758

Peters S, Michielin O, Zimmermann S.(2013) Dramatic response induced by vemurafenib in a BRAF V600E-mutated lung adenocarcinoma. J Clin Oncol. Jul 10 2013;31(20):e341-344. PMID 23733758

Petrelli F, Borgonovo K, Cabiddu M, et al.(2012) Efficacy of EGFR Tyrosine Kinase Inhibitors in Patients With EGFR-Mutated Non-Small Cell-Lung Cancer: A Meta-Analysis of 13 Randomized Trials. Clin Lung Cancer. Nov 5 2012;13(2):107-114. PMID 22056888

Pfizer.(2022) Highlights of Prescribing Information: LORBRENA (lorlatinib) tablets, for oral use. 2021; https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/210868s004lbl.pdf. Accessed October 17, 2022.

Pirker R, Pereira JR, Szczesna A, et al.(2009) Cetuximab plus chemotherapy in patients with advanced non-small-cell lung cancer (FLEX): An open-label randomized phase III trial. Lancet. 2009;373(9674):1525-1531. PMID

Planchard D, Besse B, Groen HJM, et al.(2016) Dabrafenib plus trametinib in patients with previously treated BRAF(V600E)-mutant metastatic non-small cell lung cancer: an open-label, multicentre phase 2 trial. Lancet Oncol. Jul 2016;17(7):984-993. PMID 27283860

Planchard D, Kim TM, Mazieres J, et al.(2016) Dabrafenib in patients with BRAF(V600E)-positive advanced non-small cell lung cancer: a single-arm, multicentre, open-label, phase 2 trial. Lancet Oncol. May 2016;17(5):642-650. PMID 27080216

Planchard D, Smit EF, Groen HJM, et al.(2017) Dabrafenib plus trametinib in patients with previously untreated BRAF(V600E)-mutant metastatic non-small-cell lung cancer: an open-label, phase 2 trial. Lancet Oncol. Oct 2017;18(10):1307-1316. PMID 28919011

Reck M, Rodriguez-Abreu D, Robinson AG, et al.(2016) Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N Engl J Med. Nov 10 2016; 375(19): 1823-1833. PMID 27718847

Reungwetwattana T, Nakagawa K, Cho BC, et al.(2018) CNS response to osimertinib versus standard epidermal growth factor receptor tyrosine kinase inhibitors in patients with untreated EGFR-mutated advanced non-smallcell lung cancer. J Clin Oncol. Aug 28 2018:JCO2018783118. PMID 30153097

Rizvi H, Sanchez-Vega F, La K, et al.(2018) Molecular Determinants of Response to Anti-Programmed Cell Death (PD)-1 and AntiProgrammed Death-Ligand 1 (PD-L1) Blockade in Patients With Non-Small-Cell Lung Cancer Profiled With Targeted NextGeneration Sequencing. J Clin Oncol. Mar 01 2018; 36(7): 633-641. PMID 29337640

Roberts PJ, Stinchcombe TE.(2013) KRAS mutation: should we test for it, and does it matter? J Clin Oncol. Mar 10 2013;31(8):1112-1121. PMID 23401440

Robinson SD, O'Shaughnessy JA, Cowey CL, et al.(2014) BRAF V600E-mutated lung adenocarcinoma with metastases to the brain responding to treatment with vemurafenib. Lung Cancer. Aug 2014;85(2):326-330. PMID 24888229

Rosell R, Carcereny E, Gervais R, et al.(2012) Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol. Mar 2012;13(3):239-246. PMID 2285168

Rosell R, Moran T, Queralt C, et al.(2009) Screening for epidermal growth factor receptor mutations in lung cancer. N Engl J Med. Sep 3 2009;361(10):958-967. PMID 19692684

Rudd RM, Gower NH, Spiro SG, et al.(2005) Gemcitabine plus carboplatin versus mitomycin, ifosfamide, and cisplatin in patients with stage IIIB or IV non-small-cell lung cancer: a phase III randomized study of the London Lung Cancer Group. J Clin Oncol. Jan 1 2005;23(1):142-153. PMID 15625369

Sadiq AA, Salgia R(2013) MET as a possible target for non-small-cell lung cancer. J Clin Oncol. Mar 10 2013;31(8):1089-1096. PMID 23401458

Schneider CP, Heigener D, Schott-von-Romer K, et al.(2008) Epidermal growth factor receptor-related tumor markers and clinical outcomes with erlotinib in non-small cell lung cancer: an analysis of patients from german centers in the TRUST study. J Thorac Oncol. Dec 2008;3(12):1446-1453. PMID 19057271

Schwartzberg LS, Li G, Tolba K, et al.(2022) Complementary Roles for Tissue- and Blood-Based Comprehensive Genomic Profiling for Detection of Actionable Driver Alterations in Advanced NSCLC. JTO Clin Res Rep. Sep 2022; 3(9): 100386. PMID36089920

Sequist LV, Waltman BA, Dias-Santagata D, et al.(2011) Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci Transl Med. Mar 23 2011; 3(75): 75ra26. PMID 21430269

Sequist LV, Yang JC-H, Yamamoto N, et al(2013) Phase III Study of Afatinib or Cisplatin Plus Pemetrexed in Patients With Metastatic Lung Adenocarcinoma With EGFR Mutations. J Clin Oncol. September 20, 2013 2013;31(27):3327-3334. PMID

Sezer A, Kilickap S, Gumus M, et al.(2021) Cemiplimab monotherapy for first-line treatment of advanced non-small-cell lung cancerwith PD-L1 of at least 50%: a multicentre, open-label, global, phase 3, randomised, controlled trial. Lancet. Feb 13 2021;397(10274): 592-604. PMID 33581821

Shaw AT, Bauer TM, de Marinis F, et al.(2020) First-Line Lorlatinib or Crizotinib in Advanced ALK -Positive Lung Cancer. N Engl JMed. Nov 19 2020; 383(21): 2018-2029. PMID 33207094

Shaw AT, Gandhi L, Gadgeel S, et al.(2016) Alectinib in ALK-positive, crizotinib-resistant, non-small-cell lung cancer: a single-group, multicentre, phase 2 trial. Lancet Oncol. Feb 2016;17(2):234-242. PMID 26708155

Shaw AT, Kim DW, Nakagawa K, et al.(2013) Crizotinib versus chemotherapy in advanced ALK-positive lung cancer. N Engl J Med. Jun 20 2013;368(25):2385-2394. PMID 23724913

Shaw AT, Kim TM, Crino L, et al.(2017) Ceritinib versus chemotherapy in patients with ALK-rearranged non-small-cell lung cancer previously given chemotherapy and crizotinib (ASCEND-5): a randomised, controlled, open-label, phase 3 trial. Lancet Oncol. Jul 2017;18(7):874-886. PMID 28602779

Shaw AT, Ou SH, Bang YJ, et al.(2014) Crizotinib in ROS1-rearranged non-small-cell lung cancer. N Engl J Med. Nov 20 2014;371(21):1963-1971. PMID 25264305

Shaw AT, Solomon B.(2011) Targeting anaplastic lymphoma kinase in lung cancer. Clin Cancer Res. Apr 15 2011;17(8):2081-2086. PMID 21288922

Shepherd FA, Rodrigues PJ, Ciuleanu T, et al.(2005) Erlotinib in previously treated non-small-cell lung cancer. N Engl J Med. 2005;353(2):123-132. PMID

Sim EH, Yang IA, Wood-Baker R, et al.(2018) Gefitinib for advanced non-small cell lung cancer. Cochrane Database Syst Rev. Jan 16 2018;1:CD006847. PMID 29336009

Singal G, Miller PG, Agarwala V, et al.(2019) Association of Patient Characteristics and Tumor Genomics With Clinical Outcomes Among Patients With Non-Small Cell Lung Cancer Using a Clinicogenomic Database. JAMA. Apr 09 2019; 321(14): 1391-1399. PMID 3096452

Skoulidis F, Li BT, Dy GK, et al.(2021) Sotorasib for Lung Cancers with KRAS p.G12C Mutation. N Engl J Med. Jun 24 2021; 384(25): 2371-2381. PMID 34096690

Socinski MA, Evans T, Gettinger S, et al.(2013) Treatment of stage iv non-small cell lung cancer: Diagnosis and management of lung cancer, 3rd ed: american college of chest physicians evidence-based clinical practice guidelines. Chest. 2013;143(5_suppl):e341S-e368S. PMID

Solomon BJ, Mok T, Kim DW, et al.(2014) First-line crizotinib versus chemotherapy in ALK-positive lung cancer. N Engl J Med. Dec 4 2014;371(23):2167-2177. PMID 25470694

Soria JC, Ohe Y, Vansteenkiste J, et al.(2018) Osimertinib in untreated EGFR-mutated advanced non-small-cell lung cancer. N Engl J Med. Jan 11 2018;378(2):113-125. PMID 29151359

Soria JC, Tan DSW, Chiari R, et al.(2017) First-line ceritinib versus platinum-based chemotherapy in advanced ALKrearranged non-small-cell lung cancer (ASCEND-4): a randomised, open-label, phase 3 study. Lancet. Mar 04 2017;389(10072):917-929. PMID 28126333

Sun JM, Won YW, Kim ST, et al.(2011) The different efficacy of gefitinib or erlotinib according to epidermal growth factor receptor exon 19 and exon 21 mutations in Korean non-small cell lung cancer patients. J Cancer Res Clin Oncol. Jun 16 2011;137(4):687-694. PMID 20552223

TAGRISSO (osimertinib) Prescribing Information. http://www.accessdata.fda.gov/drugsatfda_docs/label/2015/208065s000lbl.pdf.

Thunnissen E, van der Oord K, den Bakker M.(2014) Prognostic and predictive biomarkers in lung cancer. A review. Virchows Arch. Mar 2014;464(3):347-358. PMID 244207425. OSI Pharmaceuticals. Tarceva® (erlotinib) tablets for oral use prescribing information, October 2013. http://www.tarceva.com. Accessed July, 2014.

Urata Y, Katakami N, Morita S, et al.(2016) Randomized Phase III Study Comparing Gefitinib With Erlotinib in Patients With Previously Treated Advanced Lung Adenocarcinoma: WJOG 5108L J Clin Oncol. Sep 20 2016;34(27):3248-3257. PMID 27022112

Vyse S, Huang PH.(2022) Amivantamab for the treatment of EGFR exon 20 insertion mutant non-small cell lung cancer. Expert RevAnticancer Ther. Jan 2022; 22(1): 3-16. PMID 34913823

Wu D, Duan C, Wu F, et al.(2017) Which treatment is preferred for advanced non-small-cell lung cancer with wild-type epidermal growth factor receptor in second-line therapy? A meta-analysis comparing immune checkpoint inhibitor, tyrosine kinase inhibitor and chemotherapy. Oncotarget. Sep 12 2017;8(39):66491-66503. PMID 29029530

Wu YL, Saijo N, Thongprasert S, et al.(2017) Efficacy according to blind independent central review: Post hoc analyses from the phase III, randomized, multicenter, IPASS study of first-line gefitinib versus carboplatin/paclitaxel in Asian patients with EGFR mutation-positive advanced NSCLC. Lung Cancer. Feb 2017;104:119-125. PMID 28212993

Wu YL, Zhou C, Hu CP, et al.(2014) Afatinib versus cisplatin plus gemcitabine for first-line treatment of Asian patients with advanced non-small-cell lung cancer harbouring EGFR mutations (LUX-Lung 6): an open-label, randomized phase 3 trial. Lancet Oncol. Feb 2014;15(2):213-222. PMID 24439929

Wu YL, Zhou C, Liam CK, et al.(2015) First-line erlotinib versus gemcitabine/cisplatin in patients with advanced EGFR mutation-positive non-small-cell lung cancer: analyses from the phase III, randomized, open-label, ENSURE study. Ann Oncol. Sep 2015;26(9):1883-1889. PMID 26105600

Yang JC-H, Hirsh V, Schuler M, et al.(2013) Symptom Control and Quality of Life in LUX-Lung 3: A Phase III Study of Afatinib or Cisplatin/Pemetrexed in Patients With Advanced Lung Adenocarcinoma With EGFR Mutations. J Clin Oncol. September 20, 2013 2013;31(27):3342-3350. PMID

Yang JC-H, Shih J-Y, Su W-C, et al.(2012) Afatinib for patients with lung adenocarcinoma and epidermal growth factor receptor mutations (LUX-Lung 2): a phase 2 trial. Lancet Oncol. 2012;13(5):539-548. PMID

Yang JJ, Zhou Q, Yan HH, et al.(2017) A phase III randomised controlled trial of erlotinib vs gefitinib in advanced nonsmall cell lung cancer with EGFR mutations. Br J Cancer. Feb 28 2017;116(5):568-574. PMID 28103612

Yang Z, Hackshaw A, Feng Q, et al.(2017) Comparison of gefitinib, erlotinib and afatinib in non-small cell lung cancer: A meta-analysis. Int J Cancer. Jun 15 2017;140(12):2805-2819. PMID 28295308

Yang Z, Hackshaw A, Feng Q, et al.(2017) Comparison of gefitinib, erlotinib and afatinib in non-small cell lung cancer: A meta-analysis. I Int J Cancer. Jun 15 2017;140(12):2805-2819. PMID 28295308

Yoshioka H, Hotta K, Kiura K, et al.(2010) A phase II trial of erlotinib monotherapy in pretreated patients with advanced non-small cell lung cancer who do not possess active EGFR mutations: Okayama Lung Cancer Study Group trial 0705. J Thorac Oncol. Jan 2010;5(1):99-104. PMID 19898258

Yu HA, Arcila ME, Rekhtman N, et al.(2013) Analysis of tumor specimens at the time of acquired resistance to EGFR-TKI therapy in155 patients with EGFR-mutant lung cancers. Clin Cancer Res. Apr 15 2013; 19(8): 2240-7. PMID 23470965

Zhang W, Wei Y, Yu D, et al.(2018) Gefitinib provides similar effectiveness and improved safety than erlotinib for advanced non-small cell lung cancer: A meta-analysis. Medicine (Baltimore). Apr 2018;97(16):e0460. PMID 29668619

Zhang Y, Zhang Z, Huang X, et al.(2017) Therapeutic efficacy comparison of 5 major EGFR-TKIs in advanced EGFRpositive non-small-cell lung cancer: a network meta-analysis based on head-to-head trials. Clin Lung Cancer. Sep 2017;18(5):e333-e340. PMID 28462807

Zhou C, Wu YL, Chen G, et al.(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. Aug 2011;12(8):735-742. PMID 21783417

Zhu CQ, da Cunha Santos G, Ding K, et al.(2008) Role of KRAS and EGFR as biomarkers of response to erlotinib in National Cancer Institute of Canada Clinical Trials Group Study BR.21. J Clin Oncol. Sep 10 2008;26(26):4268-4275. PMID 18626007


Group specific policy will supersede this policy when applicable. This policy does not apply to the Wal-Mart Associates Group Health Plan participants or to the Tyson Group Health Plan participants.
CPT Codes Copyright © 2024 American Medical Association.