NCCN: Continuing Education
Target Audience: This activity is designed to meet the educational needs of physicians, nurses, and pharmacists involved in the management of patients with cancer.
Accreditation Statement NCCN
Physicians: National Comprehensive Cancer Network is accredited by the Accreditation Council for Continuing Medical Education (ACCME) to provide continuing medical education for physicians.
NCCN designates this journal-based CE activity for a maximum of 1.0 AMA PRA Category 1 Credit™. Physicians should claim only the credit commensurate with the extent of their participation in the activity.
Nurses: National Comprehensive Cancer Network is accredited as a provider of continuing nursing education by the American Nurses Credentialing Center‘s Commission on Accreditation.
NCCN designates this educational activity for a maximum of 1.0 contact hour.
Pharmacists: National Comprehensive Cancer Network is accredited by the Accreditation Council for Pharmacy Education as a provider of continuing pharmacy education.
NCCN designates this knowledge-based continuing education activity for 1.0 contact hour (0.1 CEUs) of continuing education credit. UAN: 0836-0000-18-007-H01-P
All clinicians completing this activity will be issued a certificate of participation. To participate in this journal CE activity: 1) review the educational content; 2) take the posttest with a 66% minimum passing score and complete the evaluation at http://education.nccn.org/node/83667; and 3) view/print certificate.
Pharmacists: You must complete the posttest and evaluation within 30 days of the activity. Continuing pharmacy education credit is reported to the CPE Monitor once you have completed the posttest and evaluation and claimed your credits. Before completing these requirements, be sure your NCCN profile has been updated with your NAPB e-profile ID and date of birth. Your credit cannot be reported without this information. If you have any questions, please e-mail education@nccn.org.
Release date: July 10, 2018; Expiration date: July 10, 2019
Learning Objectives:
Upon completion of this activity, participants will be able to:
Integrate into professional practice the updates to the NCCN Guidelines for Non–Small Cell Lung Cancer
Describe the rationale behind the decision-making process for developing the NCCN Guidelines for Non–Small Cell Lung Cancer
Disclosure of Relevant Financial Relationships
The NCCN staff listed below discloses no relevant financial relationships:
Kerrin M. Rosenthal, MA; Kimberly Callan, MS; Genevieve Emberger Hartzman, MA; Erin Hesler; Kristina M. Gregory, RN, MSN, OCN; Rashmi Kumar, PhD; Karen Kanefield; and Kathy Smith.
Individuals Who Provided Content Development and/or Authorship Assistance:
David S. Ettinger, MD, Panel Chair, has disclosed that he has served as a scientific advisor for AbbVie Inc., BeyondSpring Pharmaceuticals, Inc., Boehringer Ingelheim GmbH, Bristol-Myers Squibb Company, Eli Lilly and Company, Genentech, Inc., and Guardant Health, Inc.; and that he has received grant/research support from Golden Biotech, Inc.
Dara L. Aisner, MD, PhD, Panel Member, has disclosed that she received consulting fees/honoraria from AbbVie Inc., and Bristol-Myers Squibb Company.
Miranda Hughes, PhD, Oncology Scientist/Senior Medical Writer, NCCN, has disclosed that she has no relevant financial relationships.
This activity is supported by educational grants from AstraZeneca, Celldex Therapeutics, Celgene Corporation, Genentech, Jazz Pharmaceuticals, Inc., Novartis Pharmaceuticals Corporation, and Seattle Genetics, Inc. This activity is supported by independent educational grants from AbbVie, Merck & Co., Inc. and NOVOCURE.
Overview
Lung cancer is the leading cause of cancer death in the United States.1 In 2018, an estimated 234,030 new cases of lung and bronchial cancer will be diagnosed and 154,050 deaths will occur.2,3 However, quality of life and progression-free survival (PFS) have improved for patients with advanced non–small cell lung cancer (NSCLC) who have specific predictive biomarkers and receive targeted therapy or immunotherapy compared with those receiving chemotherapy.4–9 These NCCN Guidelines Insights focus on recent updates in targeted therapy and immunotherapy for patients with advanced NSCLC. For a list of the 2018 updates, see the complete version of the NCCN Guidelines for NSCLC (available at NCCN.org). The NCCN Guidelines for NSCLC address all aspects of disease management.
NCCN Categories of Evidence and Consensus
Category 1: Based upon high-level evidence, there is uniform NCCN consensus that the intervention is appropriate.
Category 2A: Based upon lower-level evidence, there is uniform NCCN consensus that the intervention is appropriate.
Category 2B: Based upon lower-level evidence, there is NCCN consensus that the intervention is appropriate.
Category 3: Based upon any level of evidence, there is major NCCN disagreement that the intervention is appropriate.
All recommendations are category 2A unless otherwise noted.
Clinical trials: NCCN believes that the best management for any patient with cancer is in a clinical trial. Participation in clinical trials is especially encouraged.
Biomarkers
A predictive biomarker is correlated with therapeutic efficacy based on the mechanism of action between
the therapy and the biomarker. A prognostic biomarker is indicative of patient survival independent of treatment received, because the biomarker is an indicator of the innate tumor aggressiveness. For the 2018 update, a new section was added to the NCCN Guidelines that discusses key established biomarkers and appropriate testing methods to identify them (see NSCL-G, pages 815–818).10Key established predictive biomarkers include ALK rearrangements, ROS1 rearrangements, sensitizing EGFR mutations, BRAF V600E point mutations, and PD-L1 expression levels (see NSCL-G, pages 815–818, and “Established Biomarkers,” following section). The NCCN panel recommends testing for these key established biomarkers in patients with advanced NSCLC before initial treatment, because effective targeted therapy or immunotherapy is available. The panel strongly advises broader molecular profiling to assess for emerging biomarkers (see NSCL-17, page 809, and “Emerging Biomarkers,” page 816).
Established Biomarkers
ALK Rearrangements: Approximately 5% of patients with NSCLC have ALK rearrangements; these patients tend to have adenocarcinoma histology and be never-smokers or light smokers.11,12 The NCCN panel recommends testing for ALK rearrangements (category 1 recommendation) based on the efficacy of alectinib, crizotinib, and ceritinib (see NSCL-17, page 809). Two tests have been FDA-approved for stand-alone testing or rapid prescreening: (1) a fluorescence in situ hybridization (FISH) diagnostic test, and (2) an immunohistochemistry assay.13–22 Next-generation sequencing (NGS) can be used if the platform has been appropriately designed and validated.23–27
Initial ALK-Directed Therapy: Alectinib is an oral tyrosine kinase inhibitor (TKI) that inhibits ALK rearrangements.28 A phase III randomized trial (ALEX) assessed first-line therapy with alectinib versus crizotinib in 303 patients with ALK-positive
advanced NSCLC, including those with asymptomatic central nervous system (CNS) disease.28 Disease progression or death occurred in fewer patients receiving alectinib (41% [62/152]; median follow-up of 18.6 months) compared with crizotinib (68% [102/151]; median follow-up of 17.6 months). The hazard ratio (HR) was 0.47 (95% CI, 0.34–0.65; P<.001) for disease progression or death. PFS was significantly increased with alectinib versus crizotinib: the 12-month event-free survival was 68.4% (95% CI, 61.0–75.9) compared with 48.7% (95% CI, 40.4–56.9), respectively. Median PFS was 25.7 months (95% CI, 19.9 months vs not available) for alectinib compared with 10.4 months (95% CI, 7.7–14.6 months) for crizotinib based on updated results. Fewer patients receiving alectinib had CNS progression (12%; 18/152) versus those receiving crizotinib (45%; 68/151), and response rates were 83% (126/152) versus 75% (114/151) (P=.09), respectively. Patients receiving alectinib had fewer grade 3 to 5 adverse events (AEs) compared with crizotinib (41% [63/152] vs 50% [75/151], respectively), although patients received alectinib for a longer duration than crizotinib (median, 17.9 vs 10.7 months). Two treatment-related deaths were reported for crizotinib, and none were reported for alectinib.A phase III randomized trial (J-ALEX) assessed first-line therapy with alectinib versus crizotinib in 207 Japanese patients with ALK-positive advanced NSCLC, with results similar to the ALEX trial.5 The NCCN panel recommends alectinib as a preferred first-line treatment (category 1) for ALK-positive metastatic NSCLC based on these clinical trials and FDA approval (see NSCL-21, page 810). Two other ALK inhibitors, crizotinib and ceritinib, are also recommended (category 1 for both) by the panel for first-line therapy in patients with ALK-positive NSCLC based on clinical trial data and FDA approvals.6,29,30
Subsequent Therapy: Patients typically experience disease progression after initial therapy with
alectinib, crizotinib, or ceritinib; second-line or beyond (ie, subsequent) therapy recommendations depend on the mechanism of resistance to therapy (see NSCL-22 and NSCL-23, pages 811 and 812). For patients who experience progression on first-line crizotinib, subsequent therapy for ALK-positive NSCLC includes alectinib, ceritinib, or brigatinib; continuing crizotinib is also an option, depending on the type of progression (see NSCL-22, page 811).31–35 For patients who experience progression on first-line alectinib or ceritinib, recommended subsequent therapy for ALK-positive NSCLC includes the initial cytotoxic chemotherapy regimens (eg, carboplatin/pemetrexed for nonsquamous NSCLC) used for first-line treatment in patients without genetic alterations, depending on type of progression (see NSCL-23, above).36,37 Molecular testing of a biopsy to review the resistance mechanism in this scenario may be beneficial to determine the role of other ALK inhibitors. Continuing alectinib or ceritinib may also be appropriate for some patients who experience progression on alectinib or ceritinib (see NSCL-23, above).38 In tumors with an actionable mutation, such as ALK rearrangements, immunotherapy appears to be less effective (regardless of PD-L1 expression levels) based on data in the second-line setting.39–42 Ongoing studies are assessing the role of individual ALK kinase domain mutations in selecting subsequent therapy.43,44ROS1 Rearrangements: Although ROS1 is a distinct receptor tyrosine kinase, it is very similar to ALK.45,46 ROS1 rearrangements occur in approximately 1% to 2% of patients with NSCLC, and occur more frequently in younger women (median age, 50 years) with adenocarcinoma who are never-smokers and in those negative for EGFR and KRAS mutations and ALK rearrangements.46–49 The NCCN Guidelines Panel recommends ROS1 testing based on the efficacy of crizotinib and ceritinib (see NSCL-17 and NSCL-24, pages 809 and 813).46,50,51 Testing for ROS1 rearrangements may be performed using FISH (see
NSCL-G, pages 815–818).13,48,52–54 NGS can also be used if the platform has been appropriately designed and validated.23–27,46 Immunohistochemistry testing for ROS1 requires confirmation of all positives due to a low specificity of the testing methodology. A single companion diagnostic test has been FDA-approved for ROS1 rearrangements; however, clinicians may use any appropriately validated test.51First-Line Therapy: Crizotinib inhibits ROS1 rearrangements and is FDA-approved for patients with locally advanced or metastatic ROS1-positive NSCLC.6,29,50,55–59 Crizotinib is very effective for patients with ROS1 rearrangements, yielding response rates of approximately 70% to 80%, including complete responses (CRs).46,50,51 In 50 patients with ROS1-positive advanced NSCLC, crizotinib yielded an objective response rate of 72% (95% CI, 58–84), with 3 CRs and 33 partial responses (PRs).46 The median duration of response was 17.6 months (95% CI, 14.5 months to not reached), and the median PFS was 19.2 months (95% CI, 14.4 months to not reached). Another study assessed crizotinib in 30 patients with ROS1-positive stage IV NSCLC.50 There were 5 CRs (overall response rate [ORR], 80%; disease control rate, 86.7%), and median PFS was 9.1 months. Many patients (n=26) received pemetrexed (either alone or in combination with platinum, and either before or after crizotinib) and had a response rate of 57.7% and a median PFS of 7.2 months. For the 2018 guideline update, the NCCN panel voted that crizotinib is the preferred agent for patients with ROS1-positive NSCLC based on trial data and the FDA approval (see NSCL-24, above).
Ceritinib is an oral TKI that inhibits ROS1 rearrangements.60 A phase II trial assessed ceritinib as first-line therapy in patients (n=28 evaluable) with ROS1-positive NSCLC.60 One CR and 19 PRs (ORR, 62%; 95% CI, 45%–77%) were reported. PFS was 19.3 months (95% CI, 1–37 months) for crizotinib-naïve patients and 9.3 months (95% CI, 0–22 months) for all patients; median overall survival
(OS) was 24 months (95% CI, 5–43 months). For the 2018 update, the NCCN panel recommends ceritinib (category 2A) for patients with ROS1-positive advanced NSCLC based on this trial, although it is not currently FDA-approved for this indication.Subsequent Therapy: For patients with ROS1-positive NSCLC who progress on first-line crizotinib or ceritinib, the panel recommends the initial cytotoxic chemotherapy regimens (eg, carboplatin/pemetrexed for nonsquamous NSCLC) used for first-line treatment of NSCLC in patients without genetic alterations or a clinical trial (see NSCL-24, page 813). Alectinib and ceritinib are not recommended in patients with ROS1-positive NSCLC whose disease becomes resistant to crizotinib, because no trial data suggest that these agents would be effective in this setting.46 In patients with biomarkers who have progressed on targeted therapy, immunotherapy appears to be less effective (regardless of PD-L1 expression levels).39–42 Ongoing studies are assessing new agents for patients with ROS1-positive NSCLC whose disease becomes resistant to crizotinib.61–64
BRAF V600E Mutations: BRAF is a serine/threonine kinase that is part of the MAP/ERK signaling pathway. BRAF V600E is the most common of the BRAF point mutations, occurring in 1% to 2% of patients with lung adenocarcinoma who are typically current or former smokers.65–67 BRAF mutations typically do not overlap with EGFR mutations or ALK rearrangements.65,68 The NCCN panel recommends testing for BRAF mutations based on data showing the efficacy of dabrafenib/trametinib (see NSCL-17, page 809).65,68 Real-time PCR, Sanger sequencing, and NGS are the most commonly used methods to assess for BRAF mutations (see NSCL-G, pages 815–818).
Dabrafenib and trametinib inhibit kinases in the RAS/RAF/MEK/ERK pathway.65,68 Dabrafenib inhibits BRAF harboring V600E mutations; trametinib inhibits MEK 1/2, which is downstream of BRAF signaling. A phase II trial assessed first-line
combination therapy with dabrafenib/trametinib for 36 patients with metastatic NSCLC and BRAF V600E mutations.69 The ORR was 64% (n=23; 95% CI, 46–79) with 2 CRs, and median PFS was 10.9 months (95% CI, 7.0–16.6 months). Many patients (69%; n=25) had ≥1 grade 3 or 4 AEs. Serious AEs included increased alanine aminotransferase level (14%; n=5), pyrexia (11%; n=4), increased aspartate aminotransferase level (8%; n=3), and decreased ejection fraction (8%; n=3).A phase II study assessed subsequent therapy with the dabrafenib/trametinib regimen in 57 patients with advanced NSCLC and BRAF V600E mutations whose disease had progressed on chemotherapy.65,70 The response rate was 63% (n=36) with dabrafenib/trametinib; PFS was 9.7 months (95% CI, 6.9–19.6). Serious AEs occurred in 56% of patients (n=32), including pyrexia, anemia, confusional state, hemoptysis, hypercalcemia, and cutaneous squamous cell carcinoma. Common grade 3 to 4 AEs included neutropenia in 9% (n=5), hyponatremia in 7% (n=4), and anemia in 5% (n=3). Four patients died of retroperitoneal hemorrhage, subarachnoid hemorrhage, respiratory distress, or severe disease progression. Preliminary updated data showed that patients receiving dabrafenib/trametinib had a median OS of 18.2 months (95% CI, 14.3 months to not estimable).71
Based on these trials and the FDA approval, the NCCN Guidelines Panel recommends first-line or subsequent therapy with dabrafenib/trametinib in patients with metastatic NSCLC and BRAF V600E mutations (see NSCL-25, page 814).69,71,72 First-line or subsequent chemotherapy regimens are also recommended options using the same initial cytotoxic regimens recommended for patients without genetic alterations (eg, carboplatin/pemetrexed for nonsquamous disease). Single-agent therapy with dabrafenib or vemurafenib is also an option for patients with BRAF V600E mutations who cannot tolerate combination therapy with dabrafenib/trametinib.68,71,73
Emerging Biomarkers
The NCCN Guidelines recommend broad-based molecular testing to assess for rare driver mutations for which effective drugs may be available (even if not yet FDA-approved for lung cancer), or to counsel patients regarding the availability of clinical trials in addition to assessing for established biomarkers. Emerging rare driver mutations include HER2 mutations (ie, ERBB2 mutations), RET rearrangements, high-level MET amplification, or MET exon 14 skipping mutations (see “Emerging Targeted Agents for Patients With Genetic Alterations” in the complete version of the NCCN Guidelines [NSCL-H]). Clinical trials are currently in progress for other emerging biomarkers; for example, new targeted agents are being assessed for effectiveness in patients with NTRK fusions.74,75
Immunotherapy
The NCCN panel recommends first-line pembrolizumab/carboplatin (or cisplatin)/pemetrexed (category 1) for patients with advanced nonsquamous NSCLC (ie, adenocarcinoma, large cell carcinoma) or NSCLC not otherwise specified based on data from phase II and III trials and on the FDA approval (pembrolizumab/carboplatin/pemetrexed).76,77 Pembrolizumab/chemotherapy is recommended for patients without (or unknown) genetic alterations whose PD-L1 levels are <50% or unknown. Most patients received pembrolizumab/carboplatin/pemetrexed (72%; n=445), but some received pembrolizumab/cisplatin/pemetrexed (28%; n=171); patients did not have EGFR mutations or ALK rearrangements. The estimated OS rate at 1 year was 69.2% (95% CI, 64.1–73.8) for pembrolizumab/chemotherapy versus 49.4% (95% CI, 42.1–56.2) for chemotherapy alone (HR for death, 0.49; 95% CI, 0.38–0.64; P<.001) after a median follow-up of 10.5 months. OS was improved regardless of PD-L1 expression levels, although most patients (63%) had levels of ≥1%. Median PFS was 8.8 months (95% CI, 7.6–9.2 months) for pembrolizumab/chemotherapy
versus 4.9 months (95% CI, 4.7–5.5 months) for chemotherapy alone (HR for disease progression or death, 0.52; 95% CI, 0.43–0.64; P<.001), and the response rate was 47.6% (95% CI, 42.6–52.5) versus 18.9% (95% CI, 13.8–25.0), respectively. Grade ≥3 AEs occurred at a similar rate in both arms (67.2% vs 65.8% for chemotherapy).For the 2018 update, the NCCN panel added a recommendation for atezolizumab/carboplatin/paclitaxel/bevacizumab (category 1) as first-line therapy for patients with metastatic nonsquamous NSCLC based on results of a recent phase III randomized trial (IMpower150).78 This regimen is recommended for patients whose PD-L1 levels are <50% or unknown. Maintenance therapy with atezolizumab, bevacizumab, or both is also recommended (category 1). Patients with EGFR mutations or ALK rearrangements who had experienced disease progression on (or were intolerant of) prior TKI were enrolled in this trial. Median OS was 19.2 months (95% CI, 17.0–23.8) in the atezolizumab arm compared with 14.7 months (95% CI, 13.3–16.9) in the control arm of carboplatin/paclitaxel/bevacizumab; the HR for death was 0.78 (95% CI, 0.64–0.96; P=.02). PFS was increased in the atezolizumab arm versus chemotherapy/bevacizumab (8.3 vs 6.8 months; HR, 0.62; 95% CI, 0.52–0.74; P<.001).
The NCCN panel also added a first-line therapy recommendation (category 2A) for carboplatin/paclitaxel (or nab-paclitaxel)/pembrolizumab for patients with metastatic squamous cell NSCLC based on preliminary data from the phase III KEYNOTE-407 trial.79 This pembrolizumab/chemotherapy regimen is recommended for patients whose PD-L1 levels are <50% or unknown. Maintenance therapy with pembrolizumab is also a recommended option (category 2A). Patients receiving pembrolizumab/chemotherapy had an overall response rate of 58.4% compared with 35.0% in those receiving chemotherapy alone (P=.0004).
Summary
The NCCN Guidelines for NSCLC address all aspects of disease management. These NCCN Guidelines Insights focus on recent updates to targeted therapy for patients with advanced NSCLC who have ALK or ROS1 rearrangements or BRAF V600E mutations. For the 2018 guideline update, the panel recommends crizotinib as a preferred agent for patients with ROS1 rearrangements. The panel also recommends ceritinib (category 2A) for patients with ROS1-positive advanced NSCLC. A new section was added that describes key established biomarkers and appropriate testing to identify patients with these biomarkers. The NCCN panel also revised the recommendation for pembrolizumab/carboplatin (or cisplatin)/pemetrexed to category 1 (from category 2A) based on recent trial data.78,79 In addition, the following regimens are now recommended: (1) atezolizumab/carboplatin/paclitaxel/bevacizumab (category 1) as first-line therapy for patients with metastatic nonsquamous NSCLC; and (2) pembrolizumab/carboplatin/paclitaxel (or nab-paclitaxel) (category 2A) as first-line therapy for patients with metastatic squamous cell carcinoma.
For a list of all 2018 updates, see the complete NCCN Guidelines for NSCLC (available at NCCN.org).
References
- 3.↑
Howlader N, Noone AM, Krapcho M et al.. SEER Cancer Statistics Review, 1975-2014, National Cancer Institute. Bethesda, MD. Available at: https://seer.cancer.gov/csr/1975_2014/, based on November 2016 SEER data submission. Posted April 2017.
- 4.↑
Yang JC, Wu YL, Schuler M et al.. Afatinib versus cisplatin-based chemotherapy for EGFR mutation-positive lung adenocarcinoma (LUX-Lung 3 and LUX-Lung 6): analysis of overall survival data from two randomised, phase 3 trials. Lancet Oncol 2015;16:141–151.
- 5.↑
Hida T, Nokihara H, Kondo M et al.. Alectinib versus crizotinib in patients with ALK-positive non-small-cell lung cancer (J-ALEX): an open-label, randomised phase 3 trial. Lancet 2017;390:29–39.
- 6.↑
Solomon BJ, Mok T, Kim DW et al.. First-line crizotinib versus chemotherapy in ALK-positive lung cancer. N Engl J Med 2014;371:2167–2177.
- 7.
Rosell R, Carcereny E, Gervais R et al.. 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 2012;13:239–246.
- 8.
Reck M, Taylor F, Penrod JR et al.. Impact of nivolumab versus docetaxel on health-related quality of life and symptoms in patients with advanced squamous non-small cell lung cancer: results from the CheckMate 017 study. J Thorac Oncol 2018;13:194–204.
- 9.↑
Brahmer JR, Rodriguez-Abreu D, Robinson AG et al.. Health-related quality-of-life results for pembrolizumab versus chemotherapy in advanced, PD-L1-positive NSCLC (KEYNOTE-024): a multicentre, international, randomised, open-label phase 3 trial. Lancet Oncol 2017;18:1600–1609.
- 10.↑
Lindeman NI, Cagle PT, Aisner DL et al.. 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. Arch Pathol Lab Med 2018;142:321–346.
- 11.↑
Kwak EL, Bang YJ, Camidge DR et al.. Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N Engl J Med 2010;363:1693–1703.
- 12.↑
Shaw AT, Yeap BY, Mino-Kenudson M et al.. Clinical features and outcome of patients with non-small-cell lung cancer who harbor EML4-ALK. J Clin Oncol 2009;27:4247–4253.
- 13.↑
Rogers TM, Russell PA, Wright G et al.. Comparison of methods in the detection of ALK and ROS1 rearrangements in lung cancer. J Thorac Oncol 2015;10:611–618.
- 14.
von Laffert M, Warth A, Penzel R et al.. Multicenter immunohistochemical ALK-testing of non-small-cell lung cancer shows high concordance after harmonization of techniques and interpretation criteria. J Thorac Oncol 2014;9:1685–1692.
- 15.
Wynes MW, Sholl LM, Dietel M et al.. An international interpretation study using the ALK IHC antibody D5F3 and a sensitive detection kit demonstrates high concordance between ALK IHC and ALK FISH and between evaluators. J Thorac Oncol 2014;9:631–638.
- 16.
Ali G, Proietti A, Pelliccioni S et al.. ALK rearrangement in a large series of consecutive non-small cell lung cancers: comparison between a new immunohistochemical approach and fluorescence in situ hybridization for the screening of patients eligible for crizotinib treatment. Arch Pathol Lab Med 2014;138:1449–1458.
- 17.
Zhou J, Zhao J, Sun K et al.. Accurate and economical detection of ALK positive lung adenocarcinoma with semiquantitative immunohistochemical screening. PLoS One 2014;9:e92828.
- 18.
Lindeman NI, Cagle PT, Beasley MB et al.. 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. J Thorac Oncol 2013;8:823–859.
- 19.
Thunnissen E, Bubendorf L, Dietel M et al.. EML4-ALK testing in non-small cell carcinomas of the lung: a review with recommendations. Virchows Arch 2012;461:245–257.
- 20.
Kim H, Yoo SB, Choe JY et al.. Detection of ALK gene rearrangement in non-small cell lung cancer: a comparison of fluorescence in situ hybridization and chromogenic in situ hybridization with correlation of ALK protein expression. J Thorac Oncol 2011;6:1359–1366.
- 21.
Rodig SJ, Mino-Kenudson M, Dacic S et al.. Unique clinicopathologic features characterize ALK-rearranged lung adenocarcinoma in the western population. Clin Cancer Res 2009;15:5216–5223.
- 22.↑
Mino-Kenudson M, Chirieac LR, Law K et al.. A novel, highly sensitive antibody allows for the routine detection of ALK-rearranged lung adenocarcinomas by standard immunohistochemistry. Clin Cancer Res 2010;16:1561–1571.
- 23.↑
Ali SM, Hensing T, Schrock AB et al.. Comprehensive genomic profiling identifies a subset of crizotinib-responsive ALK-rearranged non-small cell lung cancer not detected by fluorescence in situ hybridization. Oncologist 2016;21:762–770.
- 24.
Wallander ML, Geiersbach KB, Tripp SR, Layfield LJ. Comparison of reverse transcription-polymerase chain reaction, immunohistochemistry, and fluorescence in situ hybridization methodologies for detection of echinoderm microtubule-associated proteinlike 4-anaplastic lymphoma kinase fusion-positive non-small cell lung carcinoma: implications for optimal clinical testing. Arch Pathol Lab Med 2012;136:796–803.
- 25.
Weickhardt AJ, Aisner DL, Franklin WA et al.. Diagnostic assays for identification of anaplastic lymphoma kinase-positive non-small cell lung cancer. Cancer 2013;119:1467–1477.
- 26.
Jennings LJ, Arcila ME, Corless C et al.. Guidelines for validation of next-generation sequencing-based oncology panels: a joint consensus recommendation of the Association for Molecular Pathology and College of American Pathologists. J Mol Diagn 2017;19:341–365.
- 27.↑
Roy S, Coldren C, Karunamurthy A et al.. Standards and guidelines for validating next-generation sequencing bioinformatics pipelines: a joint recommendation of the Association for Molecular Pathology and the College of American Pathologists. J Mol Diagn 2018;20:4–27.
- 28.↑
Peters S, Camidge DR, Shaw AT et al.. Alectinib versus crizotinib in untreated ALK-positive non-small-cell lung cancer. N Engl J Med 2017;377:829–838.
- 29.↑
Shaw AT, Kim DW, Nakagawa K et al.. Crizotinib versus chemotherapy in advanced ALK-positive lung cancer. N Engl J Med 2013;368:2385–2394.
- 30.↑
Soria JC, Tan DS, Chiari R et al.. First-line ceritinib versus platinum-based chemotherapy in advanced ALK-rearranged non-small-cell lung cancer (ASCEND-4): a randomised, open-label, phase 3 study. Lancet 2017;389:917–929.
- 31.↑
Browning ET, Weickhardt AJ, Camidge DR. Response to crizotinib rechallenge after initial progression and intervening chemotherapy in ALK lung cancer. J Thorac Oncol 2013;8:e21.
- 32.
Camidge DR, Bang YJ, Kwak EL et al.. Activity and safety of crizotinib in patients with ALK-positive non-small-cell lung cancer: updated results from a phase 1 study. Lancet Oncol 2012;13:1011–1019.
- 33.
West H, Oxnard GR, Doebele RC. Acquired resistance to targeted therapies in advanced non-small cell lung cancer: new strategies and new agents. Am Soc Clin Oncol Educ Book 2013:272–278.
- 34.
Ou SI, Ahn JS, De Petris L et al.. Alectinib in crizotinib-refractory ALK-rearranged non-small-cell lung cancer: a phase II global study. J Clin Oncol 2016;34:661–668.
- 35.↑
Kim DW, Tiseo M, Ahn MJ et al.. Brigatinib in patients with crizotinib-refractory anaplastic lymphoma kinase-positive non-small-cell lung cancer: a randomized, multicenter phase II trial. J Clin Oncol 2017;35:2490–2498.
- 36.↑
Sandler A, Gray R, Perry MC et al.. Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. N Engl J Med 2006;355:2542–2550.
- 37.↑
Scagliotti GV, Parikh P, von Pawel J et al.. Phase III study comparing cisplatin plus gemcitabine with cisplatin plus pemetrexed in chemotherapy-naive patients with advanced-stage non-small-cell lung cancer. J Clin Oncol 2008;26:3543–3551.
- 38.↑
Ou SH, Janne PA, Bartlett CH et al.. Clinical benefit of continuing ALK inhibition with crizotinib beyond initial disease progression in patients with advanced ALK-positive NSCLC. Ann Oncol 2014;25:415–422.
- 39.↑
Borghaei H, Paz-Ares L, Horn L et al.. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med 2015;373:1627–1639.
- 40.
Brahmer J, Reckamp KL, Baas P et al.. Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med 2015;373:123–135.
- 41.
Herbst RS, Baas P, Kim DW et al.. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet 2016;387:1540–1550.
- 42.↑
Gainor JF, Shaw AT, Sequist LV et al.. EGFR mutations and ALK rearrangements are associated with low response rates to PD-1 pathway blockade in non-small cell lung cancer: a retrospective analysis. Clin Cancer Res 2016;22:4585–4593.
- 43.↑
Wei J, van der Wekken AJ, Saber A et al.. Mutations in EMT-related genes in ALK positive crizotinib resistant non-small cell lung cancers. Cancers (Basel) 2018;10:pii: E10.
- 44.↑
Gainor JF, Dardaei L, Yoda S et al.. Molecular mechanisms of resistance to first- and second-generation ALK inhibitors in ALK-rearranged lung cancer. Cancer Discov 2016;6:1118–1133.
- 45.↑
Robinson DR, Wu YM, Lin SF. The protein tyrosine kinase family of the human genome. Oncogene 2000;19:5548–5557.
- 46.↑
Shaw AT, Ou SH, Bang YJ et al.. Crizotinib in ROS1-rearranged non-small-cell lung cancer. N Engl J Med 2014;371:1963–1971.
- 47.
Dugay F, Llamas-Gutierrez F, Gournay M et al.. Clinicopathological characteristics of ROS1- and RET-rearranged NSCLC in Caucasian patients: data from a cohort of 713 non-squamous NSCLC lacking KRAS/EGFR/HER2/BRAF/PIK3CA/ALK alterations. Oncotarget 2017;8:53336–53351.
- 48.↑
Bergethon K, Shaw AT, Ou SH et al.. ROS1 rearrangements define a unique molecular class of lung cancers. J Clin Oncol 2012;30:863–870.
- 49.↑
Kim HR, Lim SM, Kim HJ et al.. The frequency and impact of ROS1 rearrangement on clinical outcomes in never smokers with lung adenocarcinoma. Ann Oncol 2013;24:2364–2370.
- 50.↑
Mazieres J, Zalcman G, Crino L et al.. Crizotinib therapy for advanced lung adenocarcinoma and a ROS1 rearrangement: results from the EUROS1 cohort. J Clin Oncol 2015;33:992–999.
- 51.↑
Kazandjian D, Blumenthal GM, Luo L et al.. Benefit-risk summary of crizotinib for the treatment of patients with ROS1 alteration-positive, metastatic non-small cell lung cancer. Oncologist 2016;21:974–980.
- 52.↑
Clave S, Gimeno J, Munoz-Marmol AM et al.. ROS1 copy number alterations are frequent in non-small cell lung cancer. Oncotarget 2016;7:8019–8028.
- 53.
Bubendorf L, Buttner R, Al-Dayel F et al.. Testing for ROS1 in non-small cell lung cancer: a review with recommendations. Virchows Arch 2016;469:489–503.
- 54.↑
Davies KD, Le AT, Theodoro MF et al.. Identifying and targeting ROS1 gene fusions in non-small cell lung cancer. Clin Cancer Res 2012;18:4570–4579.
- 55.↑
Kazandjian D, Blumenthal GM, Chen HY et al.. FDA approval summary: crizotinib for the treatment of metastatic non-small cell lung cancer with anaplastic lymphoma kinase rearrangements. Oncologist 2014;19:e5–11.
- 56.
Awad MM, Oxnard GR, Jackman DM et al.. MET exon 14 mutations in non-small-cell lung cancer are associated with advanced age and stage-dependent MET genomic amplification and c-Met overexpression. J Clin Oncol 2016;34:721–730.
- 57.
Crino L, Kim D, Riely GJ et al.. Initial phase II results with crizotinib in advanced ALK-positive non-small cell lung cancer (NSCLC): PROFILE 1005 [abstract]. J Clin Oncol 2011;29(Suppl 15):Abstract 7514.
- 58.
Camidge DR, Bang Y, Kwak EL et al.. Progression-free survival (PFS) from a phase I study of crizotinib (PF-02341066) in patients with ALK-positive non-small cell lung cancer (NSCLC) [abstract]. J Clin Oncol 2011;29(Suppl 15):Abstract 2501.
- 59.↑
Rodig SJ, Shapiro GI. Crizotinib, a small-molecule dual inhibitor of the c-Met and ALK receptor tyrosine kinases. Curr Opin Investig Drugs 2010;11:1477–1490.
- 60.↑
Lim SM, Kim HR, Lee JS et al.. Open-label, multicenter, phase II study of ceritinib in patients with non-small-cell lung cancer harboring ROS1 rearrangement. J Clin Oncol 2017;35:2613–2618.
- 61.↑
Solomon BJ, Bauer TM, Felip E et al.. Safety and efficacy of lorlatinib (PF-06463922) from the dose-escalation component of a study in patients with advanced ALK+ or ROS1+ non-small cell lung cancer (NSCLC) [abstract]. J Clin Oncol 2016;34(Suppl 15):Abstract 9009.
- 62.
Farago AF, Le LP, Zheng Z et al.. Durable clinical response to entrectinib in NTRK1-rearranged non-small cell lung cancer. J Thorac Oncol 2015;10:1670–1674.
- 63.
Zou HY, Li Q, Engstrom LD et al.. PF-06463922 is a potent and selective next-generation ROS1/ALK inhibitor capable of blocking crizotinib-resistant ROS1 mutations. Proc Natl Acad Sci U S A 2015;112:3493–3498.
- 64.↑
Katayama R, Kobayashi Y, Friboulet L et al.. Cabozantinib overcomes crizotinib resistance in ROS1 fusion-positive cancer. Clin Cancer Res 2015;21:166–174.
- 65.↑
Planchard D, Besse B, Groen HJ et al.. 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 2016;17:984–993.
- 66.
Davies H, Bignell GR, Cox C et al.. Mutations of the BRAF gene in human cancer. Nature 2002;417:949–954.
- 67.↑
Paik PK, Arcila ME, Fara M et al.. Clinical characteristics of patients with lung adenocarcinomas harboring BRAF mutations. J Clin Oncol 2011;29:2046–2051.
- 68.↑
Gautschi O, Milia J, Cabarrou B et al.. Targeted therapy for patients with BRAF-mutant lung cancer: results from the European EURAF Cohort. J Thorac Oncol 2015;10:1451–1457.
- 69.↑
Planchard D, Smit EF, Groen HJM et al.. Dabrafenib plus trametinib in patients with previously untreated BRAFV600E-mutant metastatic non-small-cell lung cancer: an open-label, phase 2 trial. Lancet Oncol 2017;18:1307–1316.
- 70.↑
Planchard D, Groen HJM, Kim TM et al.. Interim results of a phase II study of the BRAF inhibitor (BRAFi) dabrafenib (D) in combination with the MEK inhibitor trametinib (T) in patients (pts) with BRAF V600E mutated (mut) metastatic non-small cell lung cancer (NSCLC) [abstract]. J Clin Oncol 2015;33(Suppl 15):Abstract 8006.
- 71.↑
Planchard D, Besse B, Kim TM. Updated survival of patients (pts) with previously treated BRAF V600E–mutant advanced non-small cell lung cancer (NSCLC) who received dabrafenib (D) or D + trametinib (T) in the phase II BRF113928 study [abstract]. J Clin Oncol 2017;35(Suppl 15):Abstract 9075.
- 72.↑
Odogwu L, Mathieu L, Blumenthal G et al.. FDA approval summary: dabrafenib and trametinib for the treatment of metastatic non-small cell lung cancers harboring BRAF V600E mutations. Oncologist 2018;23:740–745.
- 73.↑
Planchard D, Kim TM, Mazieres J et al.. Dabrafenib in patients with BRAF(V600E)-positive advanced non-small-cell lung cancer: a single-arm, multicentre, open-label, phase 2 trial. Lancet Oncol 2016;17:642–650.
- 74.↑
Drilon A, Laetsch TW, Kummar S et al.. Efficacy of larotrectinib in TRK fusion-positive cancers in adults and children. N Engl J Med 2018;378:731–739.
- 75.↑
Drilon A, Nagasubramanian R, Blake JF et al.. A next-generation TRK kinase inhibitor overcomes acquired resistance to prior TRK kinase inhibition in patients with TRK fusion-positive solid tumors. Cancer Discov 2017;7:963–972.
- 76.↑
Gandhi L, Rodriguez-Abreu D, Gadgeel S et al.. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N Engl J Med 2018;378:2078–2092.
- 77.↑
Langer CJ, Gadgeel SM, Borghaei H et al.. Carboplatin and pemetrexed with or without pembrolizumab for advanced, non-squamous non-small-cell lung cancer: a randomised, phase 2 cohort of the open-label KEYNOTE-021 study. Lancet Oncol 2016;17:1497–1508.
- 78.↑
Socinski MA, Jotte RM, Cappuzzo F et al.. Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC. N Engl J Med 2018;378:2288–2301.
- 79.↑
Paz-Ares LG, Luft A, Tafreshi A et al.. Phase 3 study of carboplatin-paclitaxel/nab-paclitaxel (Chemo) with or without pembrolizumab for patients with metastatic squamous non–small cell lung cancer [abstract]. J Clin Oncol 2018;36(Suppl):Abstract 105.