Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 May 11.
Published in final edited form as: Am Soc Clin Oncol Educ Book. 2018 May 23;38:726–739. doi: 10.1200/EDBK_201331

Sequencing Therapy for Genetically Defined Subgroups of Non–Small Cell Lung Cancer

Helena A Yu 1, David Planchard 2, Christine M Lovly 3
PMCID: PMC10172876  NIHMSID: NIHMS1887175  PMID: 30231382

OVERVIEW

The practice of precision medicine for patients with metastatic non–small cell lung cancer (NSCLC), particularly those patients with adenocarcinoma histology (the predominant subtype of NSCLC), has become the accepted standard of care worldwide. Implementation of prospective tumor molecular profiling and rational therapeutic decision-making based on the presence of recurrently detected oncogenic “driver” alterations in the tumor genome has revolutionized the way that lung cancer is diagnosed and treated in the clinic. Over the past two decades, there has been a deluge of therapeutically actionable driver alterations and accompanying small molecule inhibitors to target these drivers. Herein, we synthesize a large and rapidly growing body of literature regarding therapeutic inhibition of driver mutations. We focus on established targets, including EGFR, anaplastic lymphoma kinase (ALK), ROS1, BRAF, RET, MET, HER2, and neurotrophic tyrosine kinase receptor (NTRK), with a particular emphasis on the sequencing of small molecule inhibitors in these genetically defined cohorts of patients with lung cancer.

SEQUENCING AGENTS IN THE TREATMENT OF PATIENTS WITH EGFR-MUTANT LUNG CANCER

EGFR-mutant lung cancers represent 12% to 17% of all lung adenocarcinomas.1,2 EGFR mutations—most commonly small deletions in exon 19 (19del) or point mutations in exon 21 (L858R)—identify a subset of patients who are most appropriately treated with first-line EGFR TKIs. Many prospective studies have compared first-line EGFR TKIs with standard cytotoxic platinum doublet chemotherapy and have confirmed the superior response rates and progression-free survival with EGFR TKIs compared with chemotherapy in patients with EGFR-mutant lung cancers.35 First-generation (erlotinib, gefitinib) and second-generation (afatinib) EGFR TKIs have multiple global approvals for first-line treatment of patients with metastatic EGFR-mutant lung cancers (Table 1). Dacomitinib (a second-generation EGFR TKI) has recently been compared head to head with gefitinib, revealing an increase in median progression-free survival (mPFS; 14.7 vs. 9.2 months, respectively).6 The results on overall survival are expected this year and could lead to a new regulatory approval.

TABLE 1.

Targeted Therapies Currently Approved and Under Development for Lung Cancer

Mutation and Drug Category/Description Approval Status
United States European Union
EGFR mutations
 Erlotinib (OSI744) First-generation EGFR TKI X X
 Gefitinib (ZD1839) First-generation EGFR TKI X X
 Afatinib (BIBW2992) Second-generation pan-HER TKI X X
 Dacomitinib (PF299804) Second-generation pan-HER TKI
 Osimertinib (AZD9291) Third-generation EGFR TKI X X
 Nazartinib (EGF816) Third-generation EGFR TKI
ALK fusion
 Crizotinib (PF2341066) First-generation ALK, MET, ROS1, multikinase inhibitor X X
 Ceritinib (LDK378) First-generation ALK, ROS1, multikinase inhibitor X X
 Alectinib (CH5424802) Second-generation ALK TKI X X
 Brigatinib (AP26113) Second-generation ALK, mutant EGFR, ROS1, multikinase inhibitor X
 Ensartinib (X396) Second-generation ALK TKI
 Lorlatinib (PF6463922) Third-generation ALK, ROS1, multikinase inhibitor
ROS1 fusion
 Crizotinib ROS1, ALK, MET, multikinase inhibitor X X
 Ceritinib ROS1, ALK, multikinase inhibitor
 Brigatinib ROS1, ALK, mutant EGFR, multikinase inhibitor
 Lorlatinib ROS1, ALK, multikinase inhibitor
 Entrectinib ROS1, ALK, TrkA/B/C (NTRK1,2,3), multikinase inhibitor
 DS-6051b ROS1, TrkA/B/C (NTRK1,2,3), multikinase inhibitor
BRAF V600E mutation
 Vemurafenib BRAF V600E selective inhibitor (serine/threonine protein kinase inhibitor)
 Dabrafenib BRAF V600E selective inhibitor (serine/threonine protein kinase inhibitor) X X
 Trametinib MEK1-2 inhibitor X X
RET fusion
 Cabozantinib RET, MET, VEGFR2, AXL, c-KIT, FLT3, multikinase inhibitor
 Vandetanib RET, EGFR, VEGFR multikinase inhibitor
 Sunitinib RET, PDGFR, VEGFR1-3, c-KIT, FLT3 multikinase inhibitor TKI
 Sorafenib RET, VEGFR1-3, PDGFR, RAF, c-KIT, FLT3 multikinase inhibitor
 Alectinib RET, ALK, FLT3 multikinase inhibitor
 Lenvatinib RET, VEGFR1-3, FGFR1-4, PDGFR multikinase inhibitor
 Nintedanib RET, VEGFR, FGFR, PDGFR, Flt3, multikinase inhibitor
 Ponatinib BCR-ABL+, FLT3, SRC, c-KIT, FGFR, VEGFR, RET multikinase inhibitor
 Regorafenib RET, VEGFR, PDGFR, FGFR, KIT, RAF multikinase inhibitor
MET amplification
 Crizotinib MET, ALK, ROS1 multikinase inhibitor
 Onartuzumab Anti-MET monoclonal antibody
 Tivantinib MET selective inhibitor
 Savolitinib MET selective inhibitor
 Glesatinib MET, AXL, multikinase inhibitor
 Capmatinib MET selective inhibitor
 Cabozantinib MET, VEGFR2, AXL, c-KIT, FLT3, RET multikinase inhibitor
MET exon 14 skipping mutation
 Crizotinib MET, ALK, ROS1 multikinase inhibitor
 Capmatinib MET selective inhibitor
NTRK fusion
 Entrectinib TrkA/B/C (NTRK1/2/3), ALK, ROS1 multikinase inhibitor
 Loxo-101 TrkA/B/C (NTRK1/2/3) TKI
 DS-6051b TrkA/B/C (NTRK1/2/3), ROS1 multikinase inhibitor
HER2 amplification
 Trastuzumab HER2 monoclonal antibody
 Ado-trastuzumab emtansine HER2 monoclonal antibody
HER mutation
 Afatinib Pan-HER TKI
 Dacomitinib EGFR, HER2 TKI
 Lapatinib EGFR, HER2 TKI
 Neratinib EGFR, HER2 TKI
 Trastuzumab HER2 monoclonal antibody
 Ado-trastuzumab emtansine HER2 monoclonal antibody

Established Treatments

These first- and second-generation EGFR TKIs are effective therapies, but the majority of patients develop disease progression on these agents after 8 to 10 months.35,7 It has become standard of care to rebiopsy patients at the time of clinical progression, and the acquired molecular alterations identified serve as the molecular mechanisms of resistance to EGFR TKI treatment.7,8 Another emerging option is liquid biopsy where tumor cfDNA within plasma is utilized for mutation testing. This has emerged as a viable alternative to tumor rebiopsy. The most common acquired mutation is EGFR T790M, but other acquired alterations include MET amplification, HER2 amplification, PIK3CA mutations, small cell histologic transformation, and epithelial to mesenchymal transition.7,8 To address these resistance mechanisms to EGFR TKIs, multiple combination treatments using first- and second-generation EGFR TKIs have been assessed. EGFR TKIs have been combined with EGFR antibodies,9,10 mTOR inhibitors,11 HDAC inhibitors,12 HSP90 inhibitors,13 MET inhibitors,14 dasatinib,15 cabozantinib,16 and other agents with limited efficacy seen with the combinations.

Osimertinib

Osimertinib is a third-generation, mutant-selective, covalent EGFR inhibitor (Table 1) that targets both the sensitizing EGFR mutations as well as EGFR T790M. Its initial approval in many countries is for patients with EGFR-mutant lung cancers who were previously treated with an EGFR TKI and have acquired EGFR T790M. This approval was based on the phase I AURA study17 and confirmed by a randomized study of osimertinib versus platinum doublet chemotherapy in this clinical setting.18 Patients treated with osimertinib as second-line EGFR TKI had an overall response rate (ORR) of 71% and an mPFS of 10.1 months. Based on the efficacy in the later-line setting, osimertinib was subsequently assessed prospectively in a randomized phase III study of osimertinib or standard of care EGFR TKI (erlotinib or gefitinib) as first-line treatment of patients with metastatic lung adenocarcinoma in the FLAURA study.19 In the first-line setting, the mPFS on osimertinib was 18.9 months compared with 10.2 months with standard EGFR TKIs. Based on these data, osimertinib is expected to receive global approval as a first-line treatment option for patients with metastatic EGFR-mutant lung cancers.

Resistance to Osimertinib and New Combination Therapies

Patients treated with osimertinib also develop resistance attributable to acquired molecular alterations. There are less robust data to suggest which mechanisms of acquired resistance occur frequently and are clinically meaningful, and we do not yet have any data on mechanisms of resistance to first-line treatment with osimertinib. Resistance mechanisms identified to third-generation EGFR TKIs include acquired EGFR C797S mutation (C797 is the site at which osimertinib binds to the EGFR kinase domain),20,21 loss of EGFR T790M,22 MET and HER2 amplification,23,24 YES1 amplification,25,26 and acquired mutations, including KRAS, PIK3CA, and HER2.24,27

New combinations will presumably be assessed both in the first-line setting and after progression on osimertinib to attempt to prevent and reverse resistance to osimertinib, respectively. The combination of osimertinib and savolitinib, an MET inhibitor, has shown activity in patients with MET amplification after EGFR TKI therapy with erlotinib, afatinib, gefitinib, or osimertinib.28 Other combinations that are being assessed include osimertinib and bevacizumab (NCT02803203, NCT03133546, and NCT02971501), osimertinib and selumetinib (NCT03392246), osimertinib and dasatinib (NCT02954523), osimertinib and the JAK inhibitor INCB039110 (NCT02917993), osimertinib and navitoclax (NCT02520778), and osimertinib and an mTOR inhibitor MLN0128 (NCT02503722) among others. With the milder toxicity profile of osimertinib compared with earlier-generation EGFR inhibitors, combination studies may prove to be more efficacious by reaching optimal doses of both drugs before being limited by toxicity.

Sequencing of EGFR Inhibitors

As a general practice, our most effective treatments should be used first, because there is a clear minority of patients who progress quickly with declining functional status and do not receive second-line therapy—as high as 50% in historical data sets.29 Other factors that should be considered when choosing a first-line treatment of this population include toxicity profile and central nervous system (CNS) efficacy. In the FLAURA study, there were fewer patients with greater than or equal to grade 3 toxicities, fewer fatal adverse events, and a lower rate of adverse events leading to permanent discontinuation with osimertinib compared with standard EGFR TKI.19 In addition, there were fewer events of CNS progression with osimertinib compared with standard EGFR TKI (6% vs. 15%). Osimertinib is well positioned to be the new first-line treatment of choice because of a marked improvement in efficacy and superior CNS penetration/efficacy while also being better tolerated by patients.

One potential criticism of first-line osimertinib is that there are no approved EGFR-directed treatments after progression on osimertinib. However, only 50% of patients on standard EGFR TKI acquire EGFR T790M and are eligible for second-line osimertinib, and the additive time on treatment with a standard EGFR TKI followed by osimertinib is essentially equivalent to the time on first-line osimertinib. After clinical progression on first-line osimertinib in the setting of C797S and the absence of EGFR T790M, there are preclinical data to suggest efficacy of a standard EGFR TKI, such as gefitinib, in that setting.30 We await clinical data that such sequencing is a viable option.

Other Strategies to Improve Outcomes for Patients With EGFR-Mutant Lung Cancers

There is substantial heterogeneity in the clinical course of patients with EGFR-mutant lung cancers. As next-generation sequencing of tumors becomes the standard of clinical care, we will be able to discern the impact of concurrent genetic alterations in addition to activating EGFR mutations within lung cancers. Several reports have shown concurrent TP53 alterations to be a negative prognostic factor31,32 associated with shorter overall survival. In addition, several pretreatment concurrent alterations, including HER2 amplification, MET amplification, and TP53 mutations, seem to be associated with shorter time to progression on EGFR TKIs.33 Highlighting concurrent alterations that may have prognostic significance is important, because it identifies a subset of patients with poorer outcomes on whom new therapeutic options should be focused.

Data from the ASPIRATION study and others have shown the utility of treatment with EGFR TKIs beyond radiographic progression in the setting of more indolent disease and in the absence of symptoms.34 Treatment beyond progression allows for additional clinical benefit and delays the time before new treatments are required. Cancers can be heterogeneous in their response and resistance to therapy, such as a scenario where the majority of the target lesions continue to respond to treatment while one lesion has begun to grow. In this situation, local therapy to the oligoprogressive metastasis followed by continuing previous systemic therapy is another means to prolong time on treatment that is largely continuing to benefit a patient.35

SEQUENCING AGENTS IN THE TREATMENT OF PATIENTS WITH ALK-REARRANGED LUNG CANCER

ALK-Rearranged Lung Cancer

Rearrangements in the gene encoding the ALK on chromosome 2p were first discovered as oncogenic driver alterations in NSCLC in 2007.36 These chromosomal rearrangements result in the production of a chimeric fusion protein—most commonly EML4-ALK, although several other ALK fusions have been described. ALK rearrangements are detected in approximately 4% to 8% of NSCLCs2,37 and can be detected in tumor samples by several diagnostic measures, including immunohistochemistry, fluorescence in situ hybridization, and next-generation sequencing.

Overview of ALK TKIs in Clinical Use

Now, approximately 10 years after the initial discovery of ALK as a lung cancer driver, there have already been numerous large, international, prospective clinical trials testing the efficacy of ALK TKIs in this patient population. Remarkably, five ALK TKIs have already gained regulatory approval (Table 1). These ALK TKIs can be broken down into three “generations” of inhibitors defined by increasing “on-target” efficacy toward ALK. Crizotinib was the first ALK TKI developed in the clinic and the first ALK TKI to obtain regulatory approval. Crizotinib also targets MET and ROS1 (described below). Therefore, the development of more potent and more specific second- and third-generation inhibitors was needed (Table 1). Below, we summarize a large amount of clinical trial data discussing the sequencing of ALK TKI therapies.

First-line Therapy for ALK-Rearranged Lung Cancer

Crizotinib was the first ALK TKI to be approved for first-line treatment of ALK+ lung cancer based on the PROFILE 1014 study.38 This phase III trial enrolled 343 patients who were treatment naïve and randomized them to crizotinib or chemotherapy (platinum/pemetrexed). Cross over to crizotinib treatment after disease progression was permitted for patients receiving chemotherapy. The ORR was 74%, mPFS was 10.9 months, and hazard ratio (HR) for progression or death in the crizotinib group was 0.45 (95% CI, 0.35–0.60). Crizotinib is approved by the U.S. Food and Drug Administration (FDA) and the European Medicines Agency for first-line therapy of ALK+ NSCLC.

More recently, ceritinib, a second-generation ALK TKI, has also been approved for first-line treatment of ALK+ lung cancer based on the ASCEND-4 study.39 This study enrolled patients with metastatic ALK+ NSCLC who were treatment naïve and had asymptomatic, neurologically stable brain metastases; 189 patients were randomly selected to receive ceritinib (750 mg orally once a day), and 187 patients were randomly selected to receive platinum/pemetrexed for four cycles (maintenance pemetrexed was allowed). Crossover to ceritinib was permitted after disease progression for patients receiving chemotherapy. mPFS was 16.6 months (95% CI, 12.6–27.2) in the ceritinib group and 8.1 months (95% CI, 5.8–11.1) in the chemotherapy group (HR 0.55; 95% CI, 0.42–0.73]; p < .00001).

In patients with measurable CNS lesions at baseline, the confirmed overall intracranial response rate was 57% (95% CI, 37%–76%) in the ceritinib arm and 22% (95% CI, 9%– 42%) in the chemotherapy arm. The most common adverse events in the ceritinib arm were diarrhea (85% all grades, 5% grade 3/4), nausea (69% all grades, 3% grade 3/4), vomiting (66% all grades, 5% grade 3/4), and an increase in liver function enzymes. The ASCEND-8 trial40 evaluated a lower dose of ceritinib (450 mg) in patients and found it to be similarly efficacious; the mPFS in the 450-mg dose arm was 17.6 months compared with 10.9 months in the 750-mg dose arm. There were also improvements with the 450- versus 750-mg dose arm for grade 3/4 diarrhea (1.1% vs. 7.8%), nausea (0% vs. 5.6%), and vomiting (0% vs. 4.4%).

Alectinib, another second-generation ALK TKI, has also been tested in the first-line setting. In the global phase III ALEX study, 286 patients with advanced/metastatic ALK+ NSCLC who were treatment naïve were enrolled and randomized to receive alectinib (600 mg orally twice a day; 152 patients) or crizotinib (250 mg orally twice a day; 151 patients).41 Platinum-based chemotherapy was not the comparator in the ALEX study. Asymptomatic brain metastases were allowed. mPFS (investigator assessment) was not reached (17.7 months to not reached) for alectinib and 11.1 months (9.1–13.1) for crizotinib, with an HR of 0.47 (95% CI, 0.34–0.65; p < .0001). For patients with baseline CNS metastases, mPFS was not reached (9.2 months to not reached) for alectinib and 7.4 months (6.6–9.6) for crizotinib, with an HR of 0.40 (95% CI, 0.25–0.64). For patients without baseline CNS metastases, mPFS was not reached for alectinib and 14.8 months (10.8–20.3) for crizotinib, with an HR of 0.51 (95% CI, 0.33–0.80). Compared with the crizotinib arm, patients in the alectinib arm experienced more myalgias (16% all grades), weight gain (10% all grades), and laboratory abnormalities, including elevated bilirubin (15% all grades) and anemia (20% all grades). Alectinib is approved by the FDA and the European Medicines Agency for first-line therapy of advanced/metastatic ALK+ NSCLC.

Several other ALK TKIs are being tested in the first-line setting. The ALTA-1L study (NCT02737501) comparing the efficacy of brigatinib versus crizotinib has completed accrual. Ensartinib (eXalt3; NCT02767804) and lorlatinib (NCT03052608) are also being tested against crizotinib in patients with treatment-naïve ALK+ lung cancer. The issue with many of these studies is that they began enrollment before the global ALEX trial was reported.

Second-Line ALK Inhibitor Therapy and Beyond

Since crizotinib was the first ALK TKI studied and approved, most of the available data to date focus on the efficacy of second- and third-generation ALK inhibitors (Table 1) in patients with acquired resistance to crizotinib. This topic has been reviewed extensively in the literature42,43 and for sake of space constraints and also relevance (with the emergence of first-line ceritinib and alectinib), will only be reviewed briefly here. In general, the factors that are important for selection of a second-line ALK TKI for a patient with acquired resistance to crizotinib are systemic activity, CNS activity, safety/adverse events profile, and resistance profiles. At present, ceritinib,4446 alectinib,47 and brigatinib48,49 are all FDA approved for patients who are intolerant of or have experienced disease progression with crizotinib. Response rates range from approximately 40% to 60% across the studies, with mPFS of approximately 5 to 15 months depending on the study. Each of these inhibitors has documented CNS activity as well.

With the emergence of second-generation inhibitors, such as ceritinib and alectinib, in the first-line setting of metastatic ALK+ NSCLC, it is not precisely clear what standard of care will emerge for second-line treatment after acquired resistance to one of these more potent ALK TKIs. It is beneficial to understand acquired resistance to these inhibitors to develop rational therapeutic strategies at the time of disease progression. As seen with other TKIs in clinical use, resistance mechanisms encompass two broad categories—“on-target” mechanisms (such as kinase domain mutations and genomic amplification of the ALK fusion) and “off-target” mechanisms (predominantly “bypass” signaling pathways). Here, we will focus on overcoming “on-target” resistance mechanisms (“off-target” resistance mechanisms will be explored in greater detail below). Although current data sets are limited, at the time of acquired resistance to second-generation ALK TKIs, ALK kinase domain mutations will be detected in approximately 50% of drug-resistant tumors. In contrast to the experience with EGFR-mutant lung cancer, where T790M is the dominant (more than 60%) resistance mutation,7,8 many different ALK resistance mutations (such as C1156Y, I1171T/N/S, F1174L/C, V1180L, R1192P, L1196M, G1202R, D1203N, S1206Y/C, E1210K, A1280V, G1269A, and others) have been described.50,51 Of these mutations, the G1202R solvent front mutation is thought to be the most recalcitrant mutation, with increasing frequency of this mutation detected with increased “on-target” potency of the ALK inhibitor.

Lorlatinib is a third-generation ALK TKI, which was designed to be increasingly selective/potent against ALK and have increased brain penetration (lorlatinib is also an ROS1 inhibitor as described in below). Lorlatinib has activity against most known ALK kinase domain mutations, including G1202R.52 Initial reports from the ongoing international phase I trial of lorlatinib (NCT01970865) have recently been published.53 Forty-one patients with ALK+ NSCLC received at least one dose of lorlatinib, 52% of whom had received two or more prior TKIs and 72% had documented CNS metastases. The ORR was 46% (95% CI, 31%–63%) for all patients who were ALK+ and 42% for those patients who had received two or more prior ALK TKIs. Adverse effects of lorlatinib included hypercholesterolemia (72% across the entire study), hypertriglyceridemia (39%), peripheral edema (39%), and peripheral neuropathy (39%). Neurocognitive adverse events (slowed speech, slowed mentation, and word-finding difficulty) were also observed, although the precise frequency was not defined.

More recently, the results from the phase II lorlatinib study were presented.54 This trial included five cohorts of patients with ALK+ NSCLC (and a sixth cohort for ROS1). In cohort 1 (30 patients who were ALK+ treatment naïve), the ORR was 90%, and the intracranial ORR was 75%. In cohorts 2 (27 patients) and 3A (32 patients; cohorts 2 and 3 included patients who were ALK+ and had previously received crizotinib only [cohort 2] or crizotinib with or without chemotherapy [cohort 3A]), the ORR was 69%, and the intracranial ORR was 68%. In cohort 3B (28 patients who were ALK+ and had previously received a noncrizotinib ALK inhibitor with or without chemotherapy), the ORR was 33%, and the intracranial ORR was 42%. In cohorts 4 (65 patients) and 5 (46 patients; cohorts 4 and 5 included patients who were ALK+ and had previously received two prior TKIs [cohort 4] or three prior TKIs [cohort 5] with/without chemotherapy), the ORR was 39%, and the intracranial ORR was 48%. These studied led the FDA to give lorlatinib breakthrough therapy designation for the treatment of patients with ALK+ metastatic NSCLC previously treated with one or more ALK inhibitors.

Rational Combination Strategies to Improve Outcomes for Patients With ALK-Rearranged Lung Cancers

Resistance to ALK TKI therapy also can be mediated by numerous ALK-independent mechanisms, most commonly thought to be activation of “bypass” signaling pathways that circumvent the inhibited ALK fusion protein. Bypass signaling pathways that have been shown to occur clinically include EGFR pathway activation,55 IGF-1R pathway activation,56 SRC signaling,57 cKIT amplification,55 and MAPK pathway activation (via KRAS copy number gain or loss of the phosphatase DUSP658). Histologic changes, such as epithelial to mesenchymal transition50 and transition to small cell lung cancer,59 have also been described. Several ongoing clinical trials hope to address the proper sequence of ALK TKIs and the role of rational combination therapies to maximize benefit and outcomes for patients with ALK+ NSCLC. For example, the combination of ceritinib and the allosteric MEK inhibitor, trametinib, is being tested in an ongoing phase I/II trial (NCT03087448). Ceritinib is also being evaluated in combination with the CDK4/6 inhibitor ribociclib/LEE011 (NCT02292550).60 Finally, ALK TKIs are being tested in combination with immune checkpoint inhibitors; however, retrospective data suggest that the magnitude of benefit seen in this cohort of patients with ALK+ NSCLC treated with checkpoint inhibitors is small.61 Additional rational study designs to forestall or overcome ALK TKI resistance are urgently needed.

OTHER TARGETABLE GENOMIC ALTERATIONS IN PATIENTS WITH LUNG CANCER

ROS1 Rearrangements

Approximately 1% of lung adenocarcinomas are driven by oncogenic ROS1 rearrangements.62 The ROS1 and ALK kinase domains show considerable homology, explaining crizotinib’s high affinity for both.63 In the phase I PROFILE 1001 study, among 50 patients with ROS1-rearranged NSCLC, the ORR was 72% with a disease control rate of 90%, and mPFS reached 19.2 months.63 In a prospective phase II study and a retrospective EUROS1 study, mPFS times were 10 and 9.1 months respectively, with ORRs of 72% and 80%, respectively.64,65 In a larger Asian phase II study, mPFS in 127 patients was 13.4 months.66 These studies led to crizotinib approval by the FDA (March 2016) and the European Medicines Agency (August 2016) for treatment of advanced ROS1-rearranged NSCLC.

Ceritinib is a second-generation ALK TKI that also has efficacy against ROS1. In a Korean phase II study, among 32 patients with ROS1 rearrangement (all crizotinib naïve except for two patients), the ORR was 67%, and mPFS reached 19.3 months; however, clinical response was not observed in the two patients who had received crizotinib.67 Other ALK TKIs—including brigatinib and lorlatinib—have shown potential anti-ROS1 activity in early development studies.68 Among three patients treated with brigatinib, the patient who was crizotinib naive had a partial response (ongoing at 21.6 months), and in 12 patients treated with lorlatinib (seven were crizotinib pretreated), the ORR was 50% with an mPFS of 7.0 months.48,53

Other potential ROS1 inhibitors include entrectinib (a potent inhibitor of ALK, ROS1 kinase, and TRK), which was evaluated in 14 patients who were crizotinib naive (13 with NSCLC and one with melanoma) in two phase I studies, giving an ORR of 86% (no responses in six patients who were pretreated with crizotinib) and an mPFS of 19 months.69 Preliminary results from a Japanese phase I study of DS-6051b (a ROS1/TRK inhibitor) gave an ORR of 62.5% among 13 patients with NSCLC (no responses in three patients who were pretreated with crizotinib).70

The most common mechanism of resistance to crizotinib in ROS1+ NSCLC is mediated by the ROS1 Gly2032Arg mutation, analogous to ALK Gly1202Arg.50 Second-generation ALK TKIs with ROS1 activity (ceritinib, brigatinib, and entrectinib) are ineffective against ROS1 Gly2032Arg. Lorlatinib retains potent activity against ROS1 Gly2032Arg in vitro and in vivo.52 Research is needed to assess its efficacy in ROS1+ patients who have relapsed after treatment with available TKIs.

BRAF Mutations

The most common BRAF mutation, V600E (Val600Glu), is observed in 1% to 2% of lung adenocarcinomas.1,71 In the phase II VE-BASKET trial with vemurafenib including patients with various BRAFV600-mutant tumors, the ORR and the mPFS in an NSCLC cohort of 19 patients were 42% and 7.3 months, respectively.72 In a recent phase II study of dabrafenib as monotherapy or combined with trametinib in patients with BRAFV600-mutant metastatic NSCLC (BRF113928), dabrafenib monotherapy gave a 33% ORR in 78 pretreated patients with a median duration of response of 9.6 months and an mPFS of 5.5 months.73,74 The BRAF-MEK TKI inhibitor combination doubled the clinical benefit in 57 pretreated patients, with an ORR of 63% and an mPFS of 10.2 months.75 The combination showed similar benefits in 36 nonpretreated patients with BRAFV600E, with an ORR of 64% and an mPFS of 10.8 months. Median overall survival was prolonged in the two combination cohorts (18.2 and 24.6 months in pretreated and nonpretreated cohorts, respectively).76 The European Medicines Agency (April 2017) and the FDA (June 2017) have approved dabrafenib in combination with trametinib for treatment of BRAFV600E-mutant advanced NSCLC.

For non–BRAFV600-mutant NSCLC, six patients received BRAF inhibitors in the retrospective study.77 All tumors with non-BRAFV600 mutant located outside the activation segment of the BRAF kinase domain (codons 596–600) were refractory to BRAF inhibitors. However, one patient with a G596V mutation achieved a partial response to vemurafenib. Additional studies are needed to assess the benefit of immune checkpoint inhibitors in patients with BRAFV600E mutants and also assess if patients with non–BRAFV600-mutant tumors can benefit from targeted therapies and/or immune checkpoint inhibitors. The current recommendation is to treat patients with BRAFV600E mutant with a BRAF-MEK inhibitor combination.

RET Rearrangements

RET fusions are found in 1% to 2% of NSCLCs and tend to be mutually exclusive with other oncogenic drivers.78 KIF5BRET is the most common, with at least 10 fusion variants. Although RET-selective TKIs have not yet been developed, several multitarget agents with anti-RET activity have been evaluated that might be restricted in their ability to inhibit RET relative to their other kinase targets. The activity of multikinase inhibitors (cabozantinib, vandetanib, sunitinib, sorafenib, alectinib, lenvatinib, nintedanib, ponatinib, and regorafenib) in RET-rearranged NSCLC (ORR = 16%–47% and mPFS = 2.3–7.3 months) is clearly inferior to that seen with selective TKIs in other oncogene-addicted NSCLC models.7981 The activity of cabozantinib in a phase II trial in RET-rearranged tumors was comparable with monotherapy BRAF TKIs in BRAFV600E mutant, with an ORR of 28% and an mPFS of 5.5 months, but clearly inferior to combination BRAF plus MEK TKIs.81 Other RET-specific TKIs are under development, and evaluation of alternative signaling pathways is needed for combination therapies to overcome RET resistance and determine the best strategies in this population.

MET Alterations

Dysregulation of the MET pathway occurs through protein overexpression, gene amplification, mutation, and rearrangement. Several agents (TKIs or monoclonal antibodies) have been developed to target MET or its ligand, hepatocyte growth factor. Early trials focused on targeting MET overexpression (15%–70% in unselected NSCLC) but without a consensus on the definition of MET positivity. MET overexpression has been particularly associated with blocking the EGFR pathway, leading to phase II/III combinations. Two randomized phase III trials failed to show any clinical benefit in OS of onartuzumab or tivantinib in association with erlotinib in unselected patients or MET protein–overexpressing NSCLC.82,83 Somatic MET mutations are diverse and include exon 14 skipping. The resulting mutant receptor shows increased MET signaling (truncated MET receptor leading to decreased ubiquitination and degradation of the MET protein). The diversity of MET exon 14 alterations presents challenges for diagnostic testing. MET exon 14 alterations are detected in 3% to 4% of NSCLCs, more frequently in adenocarcinoma and sarcomatoid histologic subtypes.84 Approximately 20% to 30% of sarcomatoid carcinomas harbor MET exon 14 alterations. Case reports and cohorts have shown dramatic and durable partial responses with MET-targeting TKIs including crizotinib, capmatinib, and cabozantinib in patients with MET exon 14. Preliminary data from the phase I trial of crizotinib (PROFILE 1001) evaluating patients with advanced lung cancer with MET exon 14 alterations showed an ORR of 44%, and global retrospective series showed an mPFS of 7 months.85 A small series has shown few responses to immunotherapy, even in patients with PD-L1 greater than or equal to 50%.86

MET amplification causes protein overexpression and constitutive kinase activation. MET copy number gains arise from two distinct processes: polysomy and amplification. They are identified by fluorescence in situ hybridization, showing an increase in the MET to CEP7 ratio, although no clear consensus on the definition of MET positivity based on gene copy number has been reached. MET amplification occurs via acquired EGFR TKI resistance, representing bypass track signaling (5%–20% of cases) or de novo (1%–5%).87 Crizotinib and capmatinib (INC280) have shown a potential clinical benefit in small phase I/II trials on MET amplification (MET/CEP7 greater than or equal to 5), with ORRs of 67% and 47%, respectively, that must be confirmed.88,89

Clinical trials focusing on combined MET and EGFR TKIs for patients with acquired resistance to EGFR TKIs are ongoing. Encouraging antitumor activity has been seen with combined osimertinib and savolitinib (a selective MET TKI) in EGFR-mutated and MET-amplified patients (confirmed centrally by fluorescence in situ hybridization; MET gene copy greater than or equal to five or MET to CEP7 ratio greater than or equal to two; the TATTON trial).28 The ORR in patients who are T790M− was 53%, and duration of response was not reached.

ERBB2/HER2 Alterations

The ERBB2 gene encoding HER2 is a major proliferative driver activating downstream signaling via the PI3K-AKT and MEK-ERK pathways. Aberrations in HER2 have emerged as oncogenic drivers and therapeutic targets in lung cancers, with HER2 mutations (exon 20) in 1% to 5% and HER2 amplifications in 2% to 5% of lung adenocarcinomas. Kinase domain mutations, mainly exon 20 insertions and point mutations, lead to constitutive HER2 kinase activation.90 Although the clinical relevance in NSCLC is questionable given the lack of definition of HER2 positivity in this indication, several case reports have shown responses with HER2 TKIs in patients with an HER2 mutation, including afatinib, lapatinib, neratinib, and neratinib plus temsirolimus.9193 Clinical benefit is generally low: for example, only three of 26 patients with HER2 mutant had a response (ORR of 12%) with dacomitinib (pan-HER TKI), 21% had a response with combined neratinib (pan-HER TKI) and temsirolimus (mTOR inhibitor), 0% had a response with neratinib alone in phase II trials, and in a recent basket trial with neratinib (SUMMIT trial), only one objective response was observed among 26 patients with lung cancer.9395 A partial response was reported in a patient harboring both an HER2 exon 20 mutation and an HER2 amplification with combined trastuzumab and paclitaxel.96 In a retrospective cohort study, ORRs of 50% and 18.2% were reported in patients with HER2 exon 20 insertions treated with combined trastuzumab-chemotherapy or afatinib, respectively, although a randomized phase II trial did not show activity with the addition of trastuzumab to chemotherapy in HER2-overexpressing lung cancer.97,98 Targeting HER2 mutations with ado-trastuzumab emtansine-1 seems promising in mutant tumors with no copy number change, with a 44% ORR, although response was low in HER2-overexpressing patients (0% ORR in IHC2+ and 20% ORR in IHC3+).99 Rarer HER2 variants include transmembrane domain mutations (e.g., V659 and G660) with sensitivity to afatinib and trastuzumab emtansine-1.99,100

NTRK Rearrangements

Among recently discovered dominant oncogenic mutations, NTRK chromosome rearrangements (less than 1%) have been identified in several solid malignancies, including NSCLC. Similar to ALK and ROS1 rearrangements, recurrent gene fusions involving NTRK1, NTRK2, and NTRK3 are actionable drivers. The rarity of these fusions across differing cancer types has resulted in basket trial design for drug development with LOXO-101 (larotrectinib) and RXDX-101 (entrectinib).69,101,102 Entrectinib is a highly potent oral ATP-competitive TKI with efficacy against TrKA (encoded by NTRK1), TrKB (encoded by NTRK2), TrKC (encoded by NTRK3), ROS1, and ALK. All three NTRK1/2/3-rearranged advanced solid tumors responded (including in one patient with NSCLC whose tumor harbored an SQSTM1–NTRK1 fusion).69 Entrectinib seems to be active in intracranial lesions and ROS1- and ALK-rearranged tumors. A phase II basket study (STARTRK-2; NCT02568267) is currently accruing patients with NTRK-, ROS1-, and ALK-rearranged cancers with the intent of confirming the results. Promising clinical benefit has also been shown with larotrectinib, a selective pan-TRK inhibitor with an ORR of 76% (95% CI, 62%–87%) in 50 patients (7% lung cancer), regardless of type of tumor and NTRK rearrangement (NTRK1/2/3).101

DISCUSSION

The rapid development of genomic biomarkers that define various molecular subtypes of NSCLC (Fig. 1) and the spectrum of currently available targeted therapies against these targets (Table 1) have completely reshaped the treatment paradigm for oncogene-addicted cancers. Nonetheless, several major challenges still remain in this field.

FIGURE 1. Molecular Cohorts of Lung Cancer.

FIGURE 1.

(A) In a large academic center, these are the actionable mutations prospectively identified on a next-generation sequencing mutation platform (430 genes) over a set time period.103 (B) In a national molecular testing effort, these are the actionable mutations (six-gene panel) prospectively identified over a set time period.1

Abbreviation: WT, wild-type.

A major challenge is making molecular testing available to the maximum number of patients. Successful implementation of personalized medicine requires widely accessible tumor molecular profiling in routine practice settings worldwide along with molecular centers for high-quality testing. Promising examples of large-scale routine molecular profiling on a national level have been reported as part of large cooperative networks in France and the United States, and they have included thousands of patients with NSCLC1,2 (Fig. 1B). It was encouraging to see that survival was improved for patients treated with biomarker-directed targeted therapies, showing the benefit of this treatment approach. Targeted gene sequencing or whole-exome sequencing by next-generation sequencing assays is gradually being integrated into clinical practice. Their success depends on them being made broadly available to practicing clinicians, applicable to small tumor biopsies, and affordable to patients and/or the health care system with a turnaround time to obtain results that is short.

An emerging option is the use of plasma genotyping with sequencing circulating tumor DNA (including the detection of high tumor mutational burden as a potential biomarker for immunotherapy), which has the logistical advantage of being rapid, noninvasive, cheap, and nononerous for the patient.104,105 It is expected to become a new standard in daily clinical practice in the near future but still needs standardization, especially for the use of a large panel of genes. By carrying out increasingly extensive molecular analyses, many uncommon or rare alterations are detected for which clinical significance assessment constitutes a real challenge. Nevertheless, the generation of massive volumes of data highlights that tools to support the medical interpretation and interaction between clinicians and scientists must be developed to ensure a thorough and rapid outcome. The implementation of molecular tumor boards both within hospitals and as national networks is increasingly widespread, offering an environment for discussion of all of these elements and dissemination of standardized practices and shared knowledge.

Another means of promoting wide access to genetic profiling is via innovative protocol designs, including molecular screening and both targeted therapy and immunotherapy arms for a single disease (umbrella trials) or a single targeted therapy for multiple diseases (basket trials). Examples of umbrella trials in advance NSCLC include the Lung-MAP (NCT02154490) for squamous cell carcinoma and the phase II trials Lung Matrix trial (NCT026649351), NCI-MATCH cooperative group trial (NCT01306045), and SAFIR02 lung trial (NCT02117167) for squamous and nonsquamous NSCLCs. In the SAFIR02 lung trial, high-throughput molecular analyses (comparative genomic hybridization array and next-generation sequencing) are used to evaluate whether treatment with guided targeted agents or immune checkpoint inhibitors improves clinical benefit compared with standard maintenance therapy in patients with metastatic NSCLC. For the basket trials, examples include the vemurafenib trial for BRAFV600-mutant patients, the French national AcSé trial (biomarker-driven access to crizotinib in ALK+, MET+, or ROS1+ malignancies in adults and children), the Larotrectinib (Loxo-101; a selective TRK inhibitor in patients with NTRK fusion cancer), and the recently published trial in HER2- and HER3-mutated patients.64,72,94,101

Another major challenge is that—despite the high response rates—all targeted therapies remain effective for a finite period of time. Improvements in outcomes for our patients will build on what we understand about how cancers escape targeted therapies (Fig. 2). One strategy is to improve on target inhibition. As has been seen in ALK+ lung cancers and EGFR-mutant lung cancers, better inhibitors can and have been developed. We have seen marked improvements in progression-free survival when we compared first-line treatment with newer agents, such as alectinib and osimertinib compared with crizotinib and erlotinib/gefitinib.19,106 Alternative dosing schedules, such as pulse dosing of targeted therapy, can be explored, especially in the setting of trying to improve CNS penetration and efficacy.107 Dual target inhibition, such as the use of afatinib and cetuximab (an EGFR TKI and an EGFR antibody, respectively), can be used for maximum on target inhibition.9,10

FIGURE 2. Mechanisms of Acquired Resistance to Targeted Therapies.

FIGURE 2.

Rational combination treatments that address known mechanisms of resistance should be assessed at the time of disease progression or at initial treatment to attempt to reverse or prevent acquired resistance. Many combinations are being studied in each of the molecular subsets of lung cancer. To truly personalize medical care, it is essential to sample the tumor at the time of acquired resistance by either tumor biopsy or analysis of plasma-derived sequencing circulating tumor DNA to identify the relevant resistance mechanisms for a particular patient. Finally, we must understand how alternative treatments, such as standard cytotoxic chemotherapy and immunotherapy, fit in when sequencing treatments for patients. There are some data to suggest that some of the oncogene-driven lung cancers are less responsive to immunotherapies,61 which makes it even more important to identify in whom and when to use immunotherapy. We also can use information regarding concurrent molecular alterations to provide both prognostic and predictive information. Although the majority of patients appropriately selected have excellent responses to targeted therapies, there are clearly outliers who have primary progression or shorter limited responses to targeted therapies. If certain concurrent alterations can identify these poor responders, we can focus efforts on developing new strategies to improve outcomes for these patients.

Overall, the prospective identification and rational therapeutic targeting of oncogenic “driver” mutations have paved the way for implementing precision treatment strategies in NSCLC and are now the standard of care worldwide. Despite much success in this area, a large amount of work is still needed to optimize the effectiveness of these therapies for patients with lung cancer and understand how to best sequence our available treatments. Collaborative efforts and integration of mutational data with multiomic, functional, and clinic-pathologic data are critical steps for the future to advance our understanding of lung oncogenesis and hopefully, turn oncogene-driven lung cancer into a chronic manageable disease state.

PRACTICAL APPLICATIONS.

  • Prospective tumor molecular profiling is now the standard of care for the treatment of patients with metastatic NSCLC. Molecular subtyping can be used to select patients for specific targeted therapeutic options.

  • Many such molecular subtypes have been described in NSCLC—including EGFR, ALK, BRAF, ROS1, RET, MET, HER2, and NTRK—and small molecule inhibitors have been developed against each of these targets.

  • The mutant-selective, third-generation EGFR TKI—osimertinib—has recently emerged as the new standard first-line therapy for metastatic EGFR-mutant NSCLC; however, it is unclear what the next best therapeutic option will be on development of osimertinib resistance. Several ongoing clinical trials are addressing combination therapies to forestall or overcome osimertinib resistance.

  • “Second-generation” ALK TKIs, including alectinib and ceritinib, have shown superior clinical outcomes for the treatment of patients with metastatic ALK+ NSCLC compared with the first-generation ALK TKI, crizotinib. These more potent inhibitors are emerging as the new standard first-line therapy for patients with metastatic ALK+ NSCLC. However, it is not clear how best to sequence ALK TKI therapy or how to overcome acquired resistance that is not mediated by an ALK kinase domain mutation.

  • Crizotinib has received regulatory approval for the management of ROS1+ NSCLCs, whereas the combination of a BRAF inhibitor (dabrafenib) and an MEK inhibitor (trametinib) has received regulatory approval for the management of BRAFV600E-mutant NSCLC.

ACKNOWLEDGMENT

H. A. Yu, D. Planchard, and C. M. Lovly contributed equally to this article.

Footnotes

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Contributor Information

Helena A. Yu, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weil Cornell Medical College, New York, NY.

David Planchard, Department of Medical Oncology, Institut Gustave Roussy, Villejuif, France.

Christine M. Lovly, Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Vanderbilt Ingram Cancer Center, Nashville, TN..

References

  • 1.Barlesi F, Mazieres J, Merlio JP, et al. ; Biomarkers France contributors. Routine molecular profiling of patients with advanced non-small-cell lung cancer: results of a 1-year nationwide programme of the French Cooperative Thoracic Intergroup (IFCT). Lancet 2016;387:1415–1426. [DOI] [PubMed] [Google Scholar]
  • 2.Kris MG, Johnson BE, Berry LD, et al. Using multiplexed assays of oncogenic drivers in lung cancers to select targeted drugs. JAMA 2014;311:1998–2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Mok TS, Wu YL, Thongprasert S, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med 2009;361:947–957. [DOI] [PubMed] [Google Scholar]
  • 4.Rosell R, Carcereny E, Gervais R, et al. ; Spanish Lung Cancer Group in collaboration with Groupe Français de Pneumo-Cancérologie and Associazione Italiana Oncologia Toracica. 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. [DOI] [PubMed] [Google Scholar]
  • 5.Sequist LV, Yang JC, Yamamoto N, et al. Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations. J Clin Oncol 2013;31:3327–3334. [DOI] [PubMed] [Google Scholar]
  • 6.Wu YL, Cheng Y, Zhou X, et al. Dacomitinib versus gefitinib as first-line treatment for patients with EGFR-mutation-positive non-small-cell lung cancer (ARCHER 1050): a randomised, open-label, phase 3 trial. Lancet Oncol 2017;18:1454–1466. [DOI] [PubMed] [Google Scholar]
  • 7.Yu HA, Arcila ME, Rekhtman N, et al. Analysis of tumor specimens at the time of acquired resistance to EGFR-TKI therapy in 155 patients with EGFR-mutant lung cancers. Clin Cancer Res 2013;19:2240–2247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Sequist LV, Waltman BA, Dias-Santagata D, et al. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci Transl Med 2011;3:75ra26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Janjigian YY, Azzoli CG, Krug LM, et al. Phase I/II trial of cetuximab and erlotinib in patients with lung adenocarcinoma and acquired resistance to erlotinib. Clin Cancer Res 2011;17:2521–2527. [DOI] [PubMed] [Google Scholar]
  • 10.Janjigian YY, Smit EF, Groen HJ, et al. Dual inhibition of EGFR with afatinib and cetuximab in kinase inhibitor-resistant EGFR-mutant lung cancer with and without T790M mutations. Cancer Discov 2014;4:1036–1045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Riely GJ, Kris MG, Zhao B, et al. Prospective assessment of discontinuation and reinitiation of erlotinib or gefitinib in patients with acquired resistance to erlotinib or gefitinib followed by the addition of everolimus. Clin Cancer Res 2007;13:5150–5155. [DOI] [PubMed] [Google Scholar]
  • 12.Reguart N, Rosell R, Cardenal F, et al. Phase I/II trial of vorinostat (SAHA) and erlotinib for non-small cell lung cancer (NSCLC) patients with epidermal growth factor receptor (EGFR) mutations after erlotinib progression. Lung Cancer 2014;84:161–167. [DOI] [PubMed] [Google Scholar]
  • 13.Johnson ML, Yu HA, Hart EM, et al. A phase I dose-escalation study of the HSP90 inhibitor AUY922 and erlotinib for patients with EGFR-mutant lung cancer with acquired resistance (AR) to EGFR tyrosine kinase inhibitors (EGFR TKIs). J Clin Oncol 2012;30:1666–1673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Rybkin II, Smit E Kopp HG, et al. PS01.60: ph Ib/II, trial of INC280 ± erlotinib vs platinum + pemetrexed in adult pts with EGFR-mutated, cMET amplified, EGFR TKI resistant, advanced NSCLC: topic: medical oncology. J Thorac Oncol 2016;11:S307–S308. [Google Scholar]
  • 15.Johnson ML, Riely GJ, Rizvi NA, et al. Phase II trial of dasatinib for patients with acquired resistance to treatment with the epidermal growth factor receptor tyrosine kinase inhibitors erlotinib or gefitinib. J Thorac Oncol 2011;6:1128–1131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Reckamp KL, Frankel HP, Mack PC, et al. Phase II trial of XL184 (cabozantinib) plus erlotinib in patients (pts) with advanced EGFR-mutant non-small cell lung cancer (NSCLC) with progressive disease (PD) on epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) therapy: a California Cancer Consortium phase II trial (NCI 9303). J Clin Oncol 2014;32:8014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Jänne PA, Yang JC, Kim DW, et al. AZD9291 in EGFR inhibitor-resistant non-small-cell lung cancer. N Engl J Med 2015;372:1689–1699. [DOI] [PubMed] [Google Scholar]
  • 18.Mok TS, Wu YL, Ahn MJ, et al. ; AURA3 Investigators. Osimertinib or platinum-pemetrexed in EGFR T790M-positive lung cancer. N Engl J Med 2017;376:629–640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Soria JC, Ohe Y, Vansteenkiste J, et al. ; FLAURA Investigators. Osimertinib in untreated EGFR-mutated advanced non-small-cell lung cancer. N Engl J Med 2018;378:113–125. [DOI] [PubMed] [Google Scholar]
  • 20.Thress KS, Paweletz CP, Felip E, et al. Acquired EGFR C797S mutation mediates resistance to AZD9291 in non-small cell lung cancer harboring EGFR T790M. Nat Med 2015;21:560–562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Yu HA, Tian SK, Drilon AE, et al. Acquired resistance of egfr-mutant lung cancer to a t790m-specific egfr inhibitor: emergence of a third mutation (c797s) in the egfr tyrosine kinase domain. JAMA Oncol 2015;1:982–984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Piotrowska Z, Niederst MJ, Karlovich CA, et al. Heterogeneity underlies the emergence of EGFRT790 wild-type clones following treatment of T790M-positive cancers with a third-generation EGFR inhibitor. Cancer Discov 2015;5:713–722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Planchard D, Loriot Y, André F, et al. EGFR-independent mechanisms of acquired resistance to AZD9291 in EGFR T790M-positive NSCLC patients. Ann Oncol 2015;26:2073–2078. [DOI] [PubMed] [Google Scholar]
  • 24.Chabon JJ, Simmons AD, Lovejoy AF, et al. Circulating tumour DNA profiling reveals heterogeneity of EGFR inhibitor resistance mechanisms in lung cancer patients. Nat Commun 2016;7:11815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ichihara E, Westover D, Meador CB, et al. SFK/FAK signaling attenuates osimertinib efficacy in both drug-sensitive and drug-resistant models of EGFR-mutant lung cancer. Cancer Res 2017;77:2990–3000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Fan P- DNG, Jayaprakash A, Venturini E, et al. YES1 amplification as a mechanism of acquired resistance (AR) to EGFR tyrosine kinase inhibitors (TKIs) identified by a transposon mutagenesis screen and clinical genomic testing. J Clin Oncol 2017;35:9043. [Google Scholar]
  • 27.Ramalingam SRT, Chewaskulyong B, Dechaphunkul A, et al. Osimertinib vs standard of care (SoC) EGFR-TKI as first-line therapy in patients (pts) with EGFRm advanced NSCLC: FLAURA. In: Proceedings of the European Society of Medical Oncology (ESMO) Madrid, Spain: European Society of Medical Oncology; 2017. Abstract LBA2_PR. [Google Scholar]
  • 28.Ahn MJ, Han J, Sequist L. TATTON phase Ib expansion cohort: osimertinib plus savolitinib for patients with EGFR-mutant MET-amplified NSCLC after progression on prior EGFR-TKI Presented at: World Conference on Lung Cancer. Yokohama, Japan; 2017. Abstract. [Google Scholar]
  • 29.Gerber DE, Rasco DW, Le P, et al. Predictors and impact of second-line chemotherapy for advanced non-small cell lung cancer in the United States: real-world considerations for maintenance therapy. J Thorac Oncol 2011;6:365–371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Niederst MJ, Engelman JA. Bypass mechanisms of resistance to receptor tyrosine kinase inhibition in lung cancer. Sci Signal 2013;6:re6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Canale M, Petracci E, Delmonte A, et al. Impact of TP53 mutations on outcome in EGFR-mutated patients treated with first-line tyrosine kinase inhibitors. Clin Cancer Res 2017;23:2195–2202. [DOI] [PubMed] [Google Scholar]
  • 32.VanderLaan PA, Rangachari D, Mockus SM, et al. Mutations in TP53, PIK3CA, PTEN and other genes in EGFR mutated lung cancers: correlation with clinical outcomes. Lung Cancer 2017;106:17–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Yu HA, Jordan E, Ni A, et al. Concurrent genetic alterations identified by next-generation sequencing in pre-treatment, metastatic EGFR-mutant lung cancers. J Clin Oncol 2016;34:9053. [Google Scholar]
  • 34.Park K, Yu CJ, Kim SW, et al. First-line erlotinib therapy until and beyond response evaluation criteria in solid tumors progression in asian patients with epidermal growth factor receptor mutation-positive non-small-cell lung cancer: the ASPIRATION study. JAMA Oncol 2016;2:305–312. [DOI] [PubMed] [Google Scholar]
  • 35.Yu HA, Sima CS, Huang J, et al. Local therapy with continued EGFR tyrosine kinase inhibitor therapy as a treatment strategy in EGFR-mutant advanced lung cancers that have developed acquired resistance to EGFR tyrosine kinase inhibitors. J Thorac Oncol 2013;8:346–351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Soda M, Choi YL, Enomoto M, et al. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature 2007;448:561–566. [DOI] [PubMed] [Google Scholar]
  • 37.Takeuchi K, Soda M, Togashi Y, et al. RET, ROS1 and ALK fusions in lung cancer. Nat Med 2012;18:378–381. [DOI] [PubMed] [Google Scholar]
  • 38.Solomon BJ, Mok T, Kim DW, et al. ; PROFILE 1014 Investigators. First-line crizotinib versus chemotherapy in ALK-positive lung cancer. N Engl J Med 2014;371:2167–2177. [DOI] [PubMed] [Google Scholar]
  • 39.Soria JC, Tan DSW, 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. [DOI] [PubMed] [Google Scholar]
  • 40.Cho BC, Kim DW, Bearz A, et al. ASCEND-8: A Randomized Phase 1 Study of Ceritinib, 450 mg or 600 mg, Taken with a Low-Fat Meal versus 750 mg in Fasted State in Patients with Anaplastic Lymphoma Kinase (ALK)-Rearranged Metastatic Non-Small Cell Lung Cancer (NSCLC). J Thorac Oncol 2017;12:1357–1367. [DOI] [PubMed] [Google Scholar]
  • 41.Peters S, Camidge DR, Shaw AT, et al. ; ALEX Trial Investigators. Alectinib versus crizotinib in untreated ALK-positive non-small-cell lung cancer. N Engl J Med 2017;377:829–838. [DOI] [PubMed] [Google Scholar]
  • 42.Caccese M, Ferrara R, Pilotto S, et al. Current and developing therapies for the treatment of non-small cell lung cancer with ALK abnormalities: update and perspectives for clinical practice. Expert Opin Pharmacother 2016;17:2253–2266. [DOI] [PubMed] [Google Scholar]
  • 43.Awad MM, Shaw AT. ALK inhibitors in non-small cell lung cancer: crizotinib and beyond. Clin Adv Hematol Oncol 2014;12:429–439. [PMC free article] [PubMed] [Google Scholar]
  • 44.Shaw AT, Kim TM, Crinò L, et al. 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 2017;18:874–886. [DOI] [PubMed] [Google Scholar]
  • 45.Kim DW, Mehra R, Tan DSW, et al. Activity and safety of ceritinib in patients with ALK-rearranged non-small-cell lung cancer (ASCEND-1): updated results from the multicentre, open-label, phase 1 trial. Lancet Oncol 2016;17:452–463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Crinò L, Ahn MJ, De Marinis F, et al. Multicenter phase II study of whole-body and intracranial activity with ceritinib in patients with ALK-rearranged non-small-cell lung cancer previously treated with chemotherapy and crizotinib: results from ASCEND-2. J Clin Oncol 2016;34:2866–2873. [DOI] [PubMed] [Google Scholar]
  • 47.Ou SH, 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. [DOI] [PubMed] [Google Scholar]
  • 48.Gettinger SN, Bazhenova LA, Langer CJ, et al. 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 2016;17:1683–1696. [DOI] [PubMed] [Google Scholar]
  • 49.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. [DOI] [PubMed] [Google Scholar]
  • 50.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Noh KW, Lee MS, Lee SE, et al. Molecular breakdown: a comprehensive view of anaplastic lymphoma kinase (ALK)-rearranged non-small cell lung cancer. J Pathol 2017;243:307–319. [DOI] [PubMed] [Google Scholar]
  • 52.Zou HY, Friboulet L, Kodack DP, et al. PF-06463922, an ALK/ROS1 inhibitor, overcomes resistance to first and second generation ALK inhibitors in preclinical models. Cancer Cell 2015;28:70–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Shaw AT, Felip E, Bauer TM, et al. Lorlatinib in non-small-cell lung cancer with ALK or ROS1 rearrangement: an international, multicentre, open-label, single-arm first-in-man phase 1 trial. Lancet Oncol 2017;18:1590–1599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Solomon B, Bauer T, 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). J Clin Oncol 2017;34:9009. [Google Scholar]
  • 55.Katayama R, Khan TM, Benes C, et al. Therapeutic strategies to overcome crizotinib resistance in non-small cell lung cancers harboring the fusion oncogene EML4-ALK. Proc Natl Acad Sci USA 2011;108:7535–7540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Lovly CM, McDonald NT, Chen H, et al. Rationale for co-targeting IGF-1R and ALK in ALK fusion-positive lung cancer. Nat Med 2014;20:1027–1034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Crystal AS, Shaw AT, Sequist LV, et al. Patient-derived models of acquired resistance can identify effective drug combinations for cancer. Science. 2014;346:1480–1486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Hrustanovic G, Olivas V, Pazarentzos E, et al. RAS-MAPK dependence underlies a rational polytherapy strategy in EML4-ALK-positive lung cancer. Nat Med 2015;21:1038–1047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Ou SI, Lee TK, Young L, et al. Dual occurrence of ALK G1202R solvent front mutation and small cell lung cancer transformation as resistance mechanisms to second generation ALK inhibitors without prior exposure to crizotinib. Pitfall of solely relying on liquid re-biopsy? Lung Cancer 2017;106:110–114. [DOI] [PubMed] [Google Scholar]
  • 60.Tripathy D, Bardia A, Sellers WR. Ribociclib (LEE011): mechanism of action and clinical impact of this selective cyclin-dependent kinase 4/6 inhibitor in various solid tumors. Clin Cancer Res 2017;23:3251–3262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Moro-Sibilot DFL, Zalcman G, Pérol M, et al. Crizotinib in patients with advanced ROS1-rearranged non-small cell lung cancer (NSCLC). Preliminary results of the ACSé phase II trial. J Clin Oncol 2015;33:8065. [Google Scholar]
  • 65.Mazières J, Zalcman G, Crinò 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. [DOI] [PubMed] [Google Scholar]
  • 66.Goto KYJ- H, Kim D-W, Lu S, et al. Phase II study of crizotinib in east Asian patients (pts) with ROS1-positive advanced non-small cell lung cancer (NSCLC). J Clin Oncol 2016;34:9022. [DOI] [PubMed] [Google Scholar]
  • 67.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. [DOI] [PubMed] [Google Scholar]
  • 68.Davare MA, Vellore NA, Wagner JP, et al. Structural insight into selectivity and resistance profiles of ROS1 tyrosine kinase inhibitors. Proc Natl Acad Sci USA 2015;112:E5381–E5390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Drilon A, Siena S, Ou SI, et al. Safety and antitumor activity of the multitargeted pan-TRK, ROS1, and ALK inhibitor entrectinib: combined results from two phase I trials (ALKA-372-001 and STARTRK-1). Cancer Discov 2017;7:400–409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Nosaki K, Fujiwara Y, Takeda M, et al. P2.06-002 phase I study of DS-6051b, a ROS1/NTRK inhibitor, in Japanese subjects with advanced solid tumors harboring either a ROS1 or NTRK fusion gene. J Thorac Oncol 2017;12:S1069 (suppl). [Google Scholar]
  • 71.Arcila ME, Drilon A, Sylvester BE, et al. MAP2K1 (MEK1) mutations define a distinct subset of lung adenocarcinoma associated with smoking. Clin Cancer Res 2015;21:1935–1943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Hyman DM, Puzanov I, Subbiah V, et al. Vemurafenib in multiple nonmelanoma cancers with BRAF V600 mutations. N Engl J Med 2015;373:726–736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Planchard D, Besse B, Groen HJM, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.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. [DOI] [PubMed] [Google Scholar]
  • 76.Planchard DBB, Kim TM, Quoix EA, et al. 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. J Clin Oncol 2017;35:9075. [Google Scholar]
  • 77.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. [DOI] [PubMed] [Google Scholar]
  • 78.Lipson D, Capelletti M, Yelensky R, et al. Identification of new ALK and RET gene fusions from colorectal and lung cancer biopsies. Nat Med 2012;18:382–384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Gautschi O, Milia J, Filleron T, et al. Targeting RET in patients with RET-rearranged lung cancers: results from the global, multicenter RET registry. J Clin Oncol 2017;35:1403–1410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Yoh K, Seto T, Satouchi M, et al. Vandetanib in patients with previously treated RET-rearranged advanced non-small-cell lung cancer (LURET): an open-label, multicentre phase 2 trial. Lancet Respir Med 2017;5:42–50. [DOI] [PubMed] [Google Scholar]
  • 81.Drilon A, Rekhtman N, Arcila M, et al. Cabozantinib in patients with advanced RET-rearranged non-small-cell lung cancer: an open-label, single-centre, phase 2, single-arm trial. Lancet Oncol 2016;17:1653–1660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Spigel DR, Edelman MJ, O’Byrne K, et al. Results from the Phase III randomized trial of onartuzumab plus erlotinib versus erlotinib in previously treated stage IIIB or IV non-small-cell lung cancer: METLung. J Clin Oncol 2017;35:412–420. [DOI] [PubMed] [Google Scholar]
  • 83.Yoshioka H, Azuma K, Yamamoto N, et al. A randomized, double-blind, placebo-controlled, phase III trial of erlotinib with or without a c-Met inhibitor tivantinib (ARQ 197) in Asian patients with previously treated stage IIIB/IV nonsquamous nonsmall-cell lung cancer harboring wild-type epidermal growth factor receptor (ATTENTION study). Ann Oncol 2015;26:2066–2072. [DOI] [PubMed] [Google Scholar]
  • 84.Frampton GM, Ali SM, Rosenzweig M, et al. Activation of MET via diverse exon 14 splicing alterations occurs in multiple tumor types and confers clinical sensitivity to MET inhibitors. Cancer Discov 2015;5:850–859. [DOI] [PubMed] [Google Scholar]
  • 85.Drilon AECD, Ou S-HI, Clark JW, et al. Efficacy and safety of crizotinib in patients (pts) with advanced MET exon 14-altered non-small cell lung cancer (NSCLC). J Clin Oncol 2016;34:108. [Google Scholar]
  • 86.Sabari JKMJ, Chen R, Dienstag JA, et al. PD-L1 expression and response to immunotherapy in patients with MET exon 14-altered non-small cell lung cancers (NSCLC). J Clin Oncol 2017;35:8512. [Google Scholar]
  • 87.Kawakami H, Okamoto I, Okamoto W, et al. Targeting MET amplification as a new oncogenic driver. Cancers (Basel) 2014;6:1540–1552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Camidge DROS- H, Shapiro G, Otterson GA, et al. Efficacy and safety of crizotinib in patients with advanced c-MET-amplified non-small cell lung cancer (NSCLC). J Clin Oncol 2014;32:8001. [Google Scholar]
  • 89.Schuler MHBR, Lim WT, Van Geel R, et al. Phase (Ph) I study of the safety and efficacy of the cMET inhibitor capmatinib (INC280) in patients (pts) with advanced cMET+ non-small cell lung cancer (NSCLC). J Clin Oncol 2016;34:9067. [Google Scholar]
  • 90.Arcila ME, Chaft JE, Nafa K, et al. Prevalence, clinicopathologic associations, and molecular spectrum of ERBB2 (HER2) tyrosine kinase mutations in lung adenocarcinomas. Clin Cancer Res 2012;18:4910–4918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.De Grève J, Teugels E, Geers C, et al. Clinical activity of afatinib (BIBW 2992) in patients with lung adenocarcinoma with mutations in the kinase domain of HER2/neu. Lung Cancer 2012;76:123–127. [DOI] [PubMed] [Google Scholar]
  • 92.Kelly RJ, Carter CA, Giaccone G. HER2 mutations in non-small-cell lung cancer can be continually targeted. J Clin Oncol 2012;30:3318–3319. [DOI] [PubMed] [Google Scholar]
  • 93.Besse BSJ- C, Yao B, Kris M. LBA39_PR neratinib (N) with or without temsirolimus (TEM) in patients (PTS) with non-small cell lung cancer (NSCLC) carrying HER2 somatic mutations: an international randomized phase II study. Ann Oncol 2014;25:mdu438.47-. [Google Scholar]
  • 94.Hyman DM, Piha-Paul SA, Won H, et al. HER kinase inhibition in patients with HER2- and HER3-mutant cancers. Nature 2018;554:189–194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Kris MG, Camidge DR, Giaccone G, et al. Targeting HER2 aberrations as actionable drivers in lung cancers: phase II trial of the pan-HER tyrosine kinase inhibitor dacomitinib in patients with HER2-mutant or amplified tumors. Ann Oncol 2015;26:1421–1427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Cappuzzo F, Bemis L, Varella-Garcia M. HER2 mutation and response to trastuzumab therapy in non-small-cell lung cancer. N Engl J Med 2006;354:2619–2621. [DOI] [PubMed] [Google Scholar]
  • 97.Mazières J, Peters S, Lepage B, et al. Lung cancer that harbors an HER2 mutation: epidemiologic characteristics and therapeutic perspectives. J Clin Oncol 2013;31:1997–2003. [DOI] [PubMed] [Google Scholar]
  • 98.Gatzemeier U, Groth G, Butts C, et al. Randomized phase II trial of gemcitabine-cisplatin with or without trastuzumab in HER2-positive non-small-cell lung cancer. Ann Oncol 2004;15:19–27. [DOI] [PubMed] [Google Scholar]
  • 99.Li BT Sr., Buonocore D, Olah ZT, et al. Ado-trastuzumab emtansine in patients with HER2 mutant lung cancers: results from a phase II basket trial. J Clin Oncol 2017;35:8510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Ou SI, Schrock AB, Bocharov EV, et al. HER2 transmembrane domain (TMD) mutations (V659/G660) that stabilize homo- and heterodimerization are rare oncogenic drivers in lung adenocarcinoma that respond to afatinib. J Thorac Oncol 2017;12:446–457. [DOI] [PubMed] [Google Scholar]
  • 101.Hyman DMLT, Kummar S, DuBois SG. The efficacy of larotrectinib (LOXO-101), a selective tropomyosin receptor kinase (TRK) inhibitor, in adult and pediatric TRK fusion cancers. J Clin Oncol 2017;35 (18_ suppl; abstr LBA2501). [Google Scholar]
  • 102.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Jordan EJ, Kim HR, Arcila ME, et al. Prospective comprehensive molecular characterization of lung adenocarcinomas for efficient patient matching to approved and emerging therapies. Cancer Discov 2017;7:596–609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Carbone DP, Reck M, Paz-Ares L, et al. ; CheckMate 026 Investigators. First-line nivolumab in stage IV or recurrent non-small-cell lung cancer. N Engl J Med 2017;376:2415–2426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Oxnard GR, Thress KS, Alden RS, et al. Association between plasma genotyping and outcomes of treatment with osimertinib (AZD9291) in advanced non-small-cell lung cancer. J Clin Oncol 2016;34:3375–3382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.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. [DOI] [PubMed] [Google Scholar]
  • 107.Yu HA, Sima C, Feldman D, et al. Phase 1 study of twice weekly pulse dose and daily low-dose erlotinib as initial treatment for patients with EGFR-mutant lung cancers. Ann Oncol 2017;28:278–284. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES