Abstract
Lung cancer is the leading cause of malignancy-related death in the United States and the second most common cancer diagnosis worldwide. In the last two decades, lung cancer treatment has evolved to include advances in the development of mutation-based targeting, immunotherapy, radiation therapy, and minimally invasive surgical techniques. The discovery of lung cancer as a molecularly heterogeneous disease has driven investigation into the development of targeted therapies resulting in improved patient outcomes. Despite these advances, there remain opportunities, through further investigation of mechanisms of resistance, to develop novel therapeutics that better direct the personalization of lung cancer therapy. In this review, we highlight developments in the evolution of targeted therapies in non-small cell lung cancer, as well as future directions shaped by emerging patterns of resistance.
Keywords: Non-small cell lung cancer, molecular biomarker, targeted therapy, oncogene, driver mutation, precision oncology
Introduction
Lung cancer remains the second-most common cancer diagnosis worldwide, afflicting 2.2 million people and accounting for 18% of cancer deaths1. In the United States (U.S.), lung cancer continues to be the leading cause of malignancy-associated death at approximately 22%2. In 2023, epidemiologists estimate that there will be 238,340 new cases of lung cancer and 127,070 deaths attributed to lung cancer in the U.S. alone3. The histopathologic subtypes of lung cancer follow the 2021 WHO classification schema and are largely based on morphology and immunohistochemical (IHC) staining patterns, which impact treatment decision-making and prognosis4. The two largest categories of lung cancer are small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC), which account for 14% and 85% of cases, respectively5. Current survival estimates of lung cancer are based on the stage at diagnosis. NSCLC has a 5-year survival rate of 65% in localized disease and 9% in metastatic disease. SCLC has even more dismal odds, with a 5-year survival rate of 30% in localized disease and 3% in metastatic disease6. In the last two decades, significant gains have been made in understanding the underlying biomolecular mechanisms that drive cancer initiation and progression, which have led to the discovery of somatic “driver mutations” (oncogenes) which, when present, block the normal regulatory cellular feedback processes that appropriately maintain cell growth and survival. Such driver mutations can transform normal cells into malignant cells characterized by unchecked growth and proliferation. Targeted therapies interrupt these aberrant pathways and have revolutionized the approach to treatment for affected patients with increased advantages of tolerability and effectiveness in advanced disease. In this review, we explore the current standard of care, as well as emerging targeted therapies in clinical trials for those with NSCLC with driver mutations; we do not address advances in chemotherapeutics or immune checkpoint inhibitors (ICIs). Within each section, we provide up-to-date epidemiological data, pathogenic mechanisms, an overview of therapeutic agents, and currently available clinical trials.
EGFR mutations
One of the most well-understood driver oncogenes in NSCLC is the EGFR (epidermal growth factor receptor) gene on chromosome 7, which encodes one of the erbB family of cell surface transmembrane receptor tyrosine kinases known as erbB-1 or HER17. Once the tyrosine kinase becomes activated, multiple downstream signaling pathways are triggered, such as the Ras-Raf-MEK-ERK, and PI3K-AKT-mTOR, promoting DNA synthesis and cell proliferation8. Dysregulation in these pathways drives tumor proliferation, differentiation, and migration. The frequency of activating mutations in EGFR in NSCLC (10–15% prevalence in the US) differs based on histology, smoking status, ethnicity, and sex. These mutations are largely observed in adenocarcinomas, those of Asian descent (22–62%), in non-smokers, and females. Multiple generations of oral EGFR tyrosine kinase inhibitors (TKIs) have been approved by the US Food and Drug Administration (FDA) for first-line therapy in those with EGFR-activating mutations in advanced NSCLC. This is a result of improved outcomes when compared to standard-of-care platinum doublet-based chemotherapy. It should be noted; however, that EGFR amplification or overexpression does not have the same predicted responsiveness to targeted therapies. More recently, testing for EGFR mutations is also recommended for those patients with resectable stage IB to IIIA NSCLC to guide consideration of adjuvant or neoadjuvant targeted therapy9.
Among those patients with EGFR driver mutations, the majority have either an exon 19 deletion (45%) or the exon 21-point mutation L858R (40%). These mutations are the most well-studied in NSCLC, with several FDA-approved therapies available impacting patient survival. The early generation TKIs (gefitinib, erlotinib, and afatinib) delivered excellent success rates in improving median progression-free survival (mPFS) when compared to standard chemotherapy in patients with EGFR mutations; however, patients develop disease progression while on therapy10–18. Osimertinib was approved by the FDA in 2018 to combat the most common resistance mechanisms, T790M mutation on exon 20 of EGFR and exon 21 L858R mutations19–22. Validation studies demonstrated superior overall survival rates for patients treated with osimertinib when compared to other EGFR-TKI therapies, particularly in those with brain metastases23,24. Unfortunately, in clinical use, we have learned that many patients develop resistance to osimertinib, which limits further definitive treatment options. Based on the current understanding of tumor heterogeneity and complex cellular signaling within the tumor and microenvironment and advances in molecular sequencing, we have identified both EGFR-dependent activating mutations and EGFR-independent mechanisms of resistance25–27.
EGFR-dependent activating mutations
A tertiary EGFR mutation, C797S, has been identified in the presence or absence of the secondary T790M mutation as playing a role in potential therapeutic options, depending on molecular positioning and availability of the binding subunits26,28–31. In vivo studies are investigating the use of brigatinib, which is a next-generation ALK-TKI, to target triple mutation (exon 19 deletion, T790M mutation, and C979S activating mutation) EGFR NSCLC32. Uncommon EGFR tertiary activating mutations on exons 18-25 include L718/G719, G796, and L792, especially when the T790M mutation is retained33,34, and S768I, S720F, E709K, and L861Q, which are typically found in conjunction with more common EGFR mutations35. Erlotinib has shown disease stability, albeit short-lived, in a subset of these patients with S786I mutations36. Afatinib is the only FDA-approved therapy for rare EGFR mutations, including S768I, L861Q, or G719X37–39. There are small signals that osimertinib has activity against these uncommon mutations in EGFR-positive NSCLC, even in patients with brain metastases40,41.
Exon 20 insertion mutations have also been discovered as potential targets for individualized therapies42,43. Due to the location on exon 20, the insertion mutation has similar properties as ERBB2 (HER2), although more heterogeneous in nature, making it difficult to systematically identify targetable features. Due to the ERBB inhibitory activity, poziotinib has been studied in vitro with good activity against exon 20 insertion mutations44. However, in early clinical studies, there are mixed results with concern for significant adverse effects of skin and gastrointestinal toxicities, which have required significant dose reduction45,46. In July 2022, Elamin et al. published a study of 50 patients with EGFR exon 20 point mutation-positive NSCLC who underwent treatment with poziotinib and achieved the primary-end point of an objective response rate (ORR) of 32% and mPFS of 5.5 months; however, clinical activity varied depending on near or far loop insertion of the point mutation47. Mobocertinib received FDA approval for subsequent breakthrough therapy in 2021 for patients with exon 20 insertion mutations after Riely et al. showed a confirmed response rate of 43%, median duration of response (mDOR) of 14 months, and mPFS of 7.3 months in 28 patients with exon 20 insertion mutations48,49. Unfortunately, the data was most promising in those without brain metastases. Significant side effects of mobocertinib included gastrointestinal side effects in up to 82% of patients with exon 20 insertion mutations. In December of 2021, in an open-label phase I/II study, Zhou et al. confirmed a durable clinical benefit for patients with previous platinum-based therapy who were treated with mobocertinib achieving an ORR of 28%, durable clinical response (DCR) of 78% and duration of response (DoR) of 17.5 months with mPFS of 7.3 months50. Amivantamab (JNJ-6118372) is a new fully human EGFR-MET antibody with an immune cell-directing activity that targets EGFR mutations, as well as MET mutations and amplifications. It inhibits ligand binding and promotes a receptor-antibody complex resulting in antibody-dependent cellular toxicity and death51. It received FDA approval for breakthrough therapy in patients with NSCLC and exon 20 insertion mutations who have already received platinum-based chemotherapy based on the CHRYSALIS study after achieving an ORR of 40%, including a complete response in 3 patients, a mDoR of 11.1 months, and mPFS of 8.3 months52. Side effects of amivantamab are usually less severe, citing rash as the most common side effect (86%), but only 4% of patients experienced severe effects. There are multiple ongoing studies testing these novel antibody mechanisms for new therapies, including the comparison of targeted therapies to the current standard of care in this rare population with limited available treatment options (NCT04129502, NCT04209465, NCT04538664)53–55. Due to significant clinical effect in other subpopulations, including those with brain metastases, osimertinib is also under investigation for patients with exon 20 insertion mutated EGFR positive NSCLC.
EGFR-independent acquired resistance mechanisms
Loss of the secondary T790M mutation usually results in EGFR-independent resistance mechanisms, which can occur via multiple pathways. Some of the most recognized to date include MET amplification, KRAS mutations, and BRAF mutations, among others33,34,56,57. MET amplification has been identified as the second most common mutation for resistance to the current EGFR-TKI therapies25,26,33,34. There are preclinical studies evaluating the use of combination EGFR/MET inhibitors in addition to third-generation EGFR-TKIs to overcome resistance with encouraging results in clinical trials in favor of combination therapy over chemotherapy58–60. Although BRAF is known as a primary oncogenic driver in NSCLC, it has also been implicated as a mechanism of resistance to EGFR-TKI therapy61–63. In preclinical studies, Jeong et al. discovered EGFR and BRAF fusion as a mechanism of resistance to the third-generation EGFR-TKI, lazertinib27. Combination therapy with lazertinib and a MEK inhibitor showed strong anti-tumor activity, suggesting a promising therapeutic option27. Some less common but still identifiable resistance mechanisms include HER2 amplification, TP53 mutation, KRAS mutation, induced epithelial-mesenchymal transformation (EMT), ALK resistance, and histologic transformation, including conversion to small cell carcinoma26,47,64–67.
Additional ERBB family mutations (HER2 mutations)
While HER2 mutations are not the next most common primary mutations in NSCLC, they are part of the ERBB receptor tyrosine kinase family and are studied in EGFR mutant NSCLC. HER2 mutations are present in 2–4% of NSCLC and typically expressed in adenocarcinoma among never smokers and women with a reported incidence of 20.4%68–70. HER2-positive status in NSCLC appears to confer resistance to a standard regimen of platinum-based chemotherapy70,71. There are three categories of inhibitors, including pan-HER TKIs, anti-HER2 monoclonal antibodies, and anti-HER2/HER3 antibody-drug conjugates, that have been used to investigate potential targeted treatment options for those with NSCLC72,73. Patritumab (HER3-DXd) is an antibody-drug conjugate consisting of a HER3 antibody attached to a topoisomerase I inhibitor. This molecule has been studied in Phase I trials in patients with EGFR-mutated NSCLC who developed resistance to EGFR-TKI therapy, with an ORR of 39% and mPFS of 8.2 months74. Poziotinib and afatinib showed mixed results in a study of 7 patients, mostly consisting of women non-smokers with stage IV adenocarcinoma and no coexisting EGFR or ALK mutations68. The NICHE study reported mixed results in which afatinib achieved disease control in less than 50% of the study participants at 12 weeks75. In 2021, Li et al. reported that trastuzumab deruxtecan achieved ORR of 55%, mDoR of 9.3 months, mPFS of 8.2 months, and median overall survival (mOS) of 17.8 months in patients with HER2-mutated NSCLC (DESTINY-Lung01)76. The authors of the DESTINY-Lung 02 trial reported a confirmed ORR of 56% and mDOR of 8.7 months, which gained trastuzumab deruxtecan accelerated approval by the FDA in August 2022 for patients with HER2-mutated NSCLC77. While there was an adequate safety profile, a select number of patients developed interstitial lung disease, resulting in 2 deaths76. More investigation is required to determine additional therapy options.
Studies are ongoing to investigate the emerging resistance patterns of EGFR-mutant NSCLC and new potential therapies78–81. Although many molecular mechanisms of resistance to EGFR-TKI therapy have been identified, our understanding of how these mechanisms impact therapeutic effects is incomplete.
RAS mutations
The KRAS proto-oncogene drives several hallmarks of cancer, including cellular proliferation, differentiation, and survival. KRAS mutations are commonly found in about 30% of patients with adenocarcinoma82–84. However, selective targeting of this mutation remains a significant challenge due to multiple downstream signaling pathways. The most common KRAS mutation is KRAS-G12C which is present in about 13% of lung adenocarcinomas. In August of 2020, Jänne et al. reported the success of adagrasib in the KRYSTAL-1 study, where 45% of 51 patients with NSCLC had an objective response and DCR of 96% with mDoR of 8.5 months, which ultimately led to FDA approval of adagrasib in December 202285. Preclinical animal model data supports the need for ongoing studies for the use of adagrasib in brain metastases86. In May of 2021, the FDA-approved sotorasib, a RAS GTPase inhibitor, for treatment in those with KRAS G12C mutated locally advanced or metastatic NSCLC who have received at least one prior systemic therapy87. Approval was based on the results of the Codebreak 100 trial, which showed significant responses (37% ORR and 3.2% of patients achieving complete response) with a favorable safety profile and mPFS of 6–8 months87–89. As with other kinase inhibitors, some patients are now developing resistance with evidence of disease progression while on therapy. The mechanisms of resistance remain incompletely understood. Awad et al. described 38 patients with KRAS G12C mutations, 10 of whom had colorectal cancer, 1 with appendiceal cancer, and the remaining 27 with NSCLC, who all progressed on adagrasib after 12 weeks90. 84% of the patients maintained the original KRAS G12C mutation while on therapy. The remaining 16% had no detectable KRAS G12C mutation in plasma after therapy. Mechanisms of resistance included novel secondary KRAS mutations (Y96C, H95Q, H95R, R68S, H95D) in four patients, in addition to KRAS-activating mutations (G12D, G12V, G13D, G12W)90. Pathogenic mutations in other receptor tyrosine kinase (RTK)-RAS MAPK pathways were also detected, including NRAS, BRAF-V600E, MAP2K1/MEK1, EGFR, histological transformation, and high-level focal amplification in the KRAS G12C allele, in addition to acquired MET amplification90,91. PI3K-AKT has also been implicated as a likely resistance pattern for KRAS-targeted therapies92. Deep mutational scanning was used to identify all possible alleles with a single amino acid substitution within KRAS G12C. Novel drug resistance mutations were identified at codons 8, 9, 12, 64, 68, 95, 96, 99, and 117 with strong resistance to KRAS G12C inhibitors in addition to the known adagrasib-resistant KRAS mutations, which were also resistant to MRTX1257 (sotorasib analog)90. There were also novel strong resistance mutations to sotorasib at codons 8, 9, 12, 96, and 11790. Most were commonly found to occur at the drug-binding site; however, there were amino acids located outside of the drug-binding pocket that impeded GTP hydrolysis (G13D and Q16R) or facilitated GDP to GTP nucleotide exchange (G13D, A59S, and A146P)90. Several highly selective KRAS G12C inhibitors are in development, including those that bind to the active GTP bound conformation, which in theory, increases the inhibitory activity and promotes control (RMC-6261)91. Epithelial mesenchymal transition (EMT) has been shown to lead to both acquired and intrinsic resistance to KRAS G12C inhibition with multi-targeted therapy showing tumor regression in mice models93. There are studies underway to identify additional targets in these pathways94,95. In addition to the approval of sotorasib and adagrasib, there are many more KRAS G12C pathway inhibitors under investigation83,96–101. There are also active studies of the precursor signals that activate KRAS and contribute to cell survival and proliferation, like Son of Sevenless (SOS1) and SHP2, which have the potential to overcome acquired resistance to KRAS-targeted therapies102–108.
MET mutations or amplification
MET alterations, most commonly skip mutations, occur in 3–4% of NSCLC, and drive tumor proliferation, invasion, and metastasis10. A nonselective MET inhibitor, crizotinib achieved an ORR of 32% and mDoR of 9.1 months with PFS of 7.3 months and has been validated in follow-up phase II studies in patients with MET exon 14 mutations109,110. Crizotinib was approved for breakthrough therapy in this subclass of patients by the FDA in 2018. In a phase II study, Wolf et al. proved that capmatinib exhibited an ORR of 41% in 69 patients who had previously received therapy and an ORR of 68% in treatment naïve patients111. The mDoR was 9.7 months and 12.6 months, respectively. There was less of a response in patients with MET amplification mutations, with an ORR of 29% and 40% for those who were previously treated and treatment naïve, respectively111. There are phase II studies reporting activity and response with tepotinib, savolitinib, and capmatinib in NSCLC patients harboring MET exon 14 skip mutations111–113. The only FDA-approved MET-targeted therapies include capmatinib, which was approved in May 2020, and tepotinib, approved in February 2021114,115. Phase III trials are ongoing comparing capmatinib to chemotherapy in those with MET exon 14 skip mutations in NSCLC116. The mechanisms of resistance to early-generation MET-TKIs are not fully understood but found to include many of the same resistance mechanisms described in EGFR-positive NSCLC. Recondo et al. studied 20 patients and identified genetic alterations or bypass signaling in 15 of those patients, which included single and polyclonal MET kinase domain mutations at codons H1094, G1163, L1195, D1228, Y1230, high levels of MET amplifications on exon 14, KRAS mutations and amplifications, EGFR, HER3, and BRAF117. There are small case reports of BRAF V600E mutation leading to resistance to crizotinib118.
BRAF mutations
BRAF is a RAS-activated intracellular serine/tyrosine kinase protein. BRAF mutations are identified in about 2–5% of NSCLCs, resulting in persistent downstream signaling of mitogen-activated protein kinase (MAPK), driving tumor growth and proliferation10. Activating V600E mutations comprise half of the cases of BRAF mutations119,120. Resistance to therapy emerges through MEK 1 and 2 signaling pathways121–123. There are three types of targeting inhibitors under investigation (BRAF inhibitors, MEK 1 and 2 inhibitors, and ERK inhibitors). BRAF inhibitor therapy alone has little effect as monotherapy121,124. However, the BRAF inhibitor (dabrafenib) plus MEK1 and 2 inhibitor (trametinib) demonstrate good activity in phase II clinical trials with an ORR of 63% and mDoR of 9 to 15 months125–127. The BRAF inhibitor vemurafenib demonstrates activity among those with BRAF-V600E mutations exhibiting ORR between 37–44% and mPFS of 6.5 months128,129. Currently, the only FDA-approved regimen for patients with BRAF V600E mutations in NSCLC is the combination of two agents, trametinib and dabrafenib130. Co-occurring mutations most commonly include TP53 in small sample sizes131. Unfortunately, very little is known about the resistance mechanisms to BRAF inhibitor therapy132. There are multiple drugs under investigation for the treatment of BRAF-mutant NSCLC133–136.
ALK rearrangements and ROS1 rearrangements
ALK fusion mutations drive tumor cell proliferation and survival and are present in 4–5% of adenocarcinomas137–139. ROS 1 is a kinase receptor with significant structural similarity to ALK10. Rearrangements in ROS 1 can develop into fusions of the ROS tyrosine kinase domain with other genes which occur in about 1–2% of NSCLC121. There are two main inhibitors associated with ROS, those that inhibit ROS1 and ALK and those that inhibit ROS1 and tyrosine receptor kinase (TRK). Crizotinib, in addition to MET inhibition, also inhibits ALK/ROS1, which was FDA approved in 2011 after early studies showed promising results for patients with fusion mutations and validated in later phase III studies140–144. To date, the ALK inhibitors ceritinib, alectinib, and brigatinib show improved ORR and mPFS when compared to either standard chemotherapy or crizotinib alone in treatment naïve patients145–147. Lorlatinib is a selective ALK/ROS1 TKI with brain penetration. In 2020, Shaw et al. compared lorlatinib to crizotinib and discovered improved overall survival (OS) at 12 months in the lorlatinib group – 78% to 39%, respectively, an ORR of 76% and 58%, respectively, and brain metastasis response rate of 82% in lorlatinib and 23% in crizotinib144. Lorlatinib received FDA approval for a breakthrough therapy for a DCR rate of 50% overall, including intracranial response in those with brain metastases. In 2021, Camidge et al. reported the benefits of brigatinib as first-line therapy in patients with ALK inhibitor naïve -ALK-positive NSCLC, with superior 3-year PFS versus crizotinib, and increased survival in those with brain metastases148. TP53 mutations are the most common genomic co-alteration in ALK-positive NSCLC, with overall worse prognosis and survival, with preclinical studies testing ixazomib (a proteasome inhibition), which induced apoptosis in previously alectinib resistance cells149. Known mechanisms of resistance to ALK-TKI therapy include secondary ALK mutations, BRAF-V600E mutations, and a novel discovery of epithelial-mesenchymal transition (EMT) of newly metastatic cells150. Three additional ROS1/TRK inhibitors have been studied in ROS1-rearranged NSCLC. Entrectinib is a highly potent ATP-competitive TKI that penetrates the central nervous system. Multiple studies show an ORR of 67.1%, DoR of 15.7 months, and mPFS of 15.7 months in 161 TKI naïve patients151. Repotrectinib and taletrectinib are ROS1/TRK inhibitors that can penetrate the CNS. The first in-human phase I study of taletrectinib achieved an ORR of 33.3% in those with critzotinib resistance ROS1 mutations152. Repotrectinib has shown activity against ROS1-resistance mutations and there are ongoing studies to confirm anti-tumor activity153. Entrectinib is the only ROS1/TRK inhibitor to be approved by the FDA in 2019151,154,155.
RET rearrangements
The RET gene encodes a cell surface tyrosine kinase receptor that impacts cell proliferation and differentiation. In 1–2% of those with NSCLC (most often in adenocarcinomas, never-smokers, and younger patients), rearrangements between RET and CCDC6 (coiled-coil domain containing-6), KIF5B (kinesin family 5B), NCOA4 (nuclear receptor coactivator 4), or other domains ultimately results in dysregulated signaling and RET overexpression. Testing via next-generation sequencing (NGS) (RNA-based preferred over DNA-based testing) is the preferred method. Break-apart FISH (fluorescence in situ hybridization) probes or RT-PCR (reverse transcription polymerase chain reaction) can be used; however, they may not detect less common fusion products. Regardless of performance status, first-line systemic therapy for those patients with advanced NSCLC (even with brain metastases) and a RET fusion is often with either of the oral RET TKIs (selpercatinib or pralsetinib), especially since current evidence is limited and suggests mixed efficacy of immune checkpoint inhibitor monotherapy with an ORR of 6%156–158. The landmark phase I/II multi-cohort open-label trial that led to FDA approval for selpercatinib was LIBRETTO-00, where patients were either treatment-naïve or previously received platinum-based chemotherapy for their RET fusion NSCLC. In the 69 treatment-naïve patients, an ORR of 84%, mPFS of 22 months, and mDoR of 20 months were reported, whereas the 247 previously treated patients had an ORR of 61%, mPFS of 25 months, and mDoR of 29 months159,160. Among those patients with brain metastases, 26 had baseline measurable disease and were without any radiation therapy in the two months preceding trial enrollment, and 22 patients had an intracranial response (ORR of 85%) with mDoR of 9.4 months; this was consistent whether prior local or systemic treatments were received160,161. These rearrangements result in ligand-independent signaling, which leads to oncogenic proliferation and has been implicated in the pattern of resistance to osimertinib121. Small studies investigating multi-kinase inhibitors like cabozantinib and vandetinib have reported ORR of 28% and 47%, respectively, in NSCLC162,163. In 2020, more selective RET inhibitors such as pralsetinib demonstrated promising results achieving ORR of 65%, time to response of 1.8 months, and mDoR not being met164. Selpercatinib was tested in 105 patients with RET fusion-positive NSCLC and achieved an ORR of 64% and DoR of 17.5 months159. The results were superior for treatment-naïve patients achieving an ORR of 85%, and those with nervous system symptoms with an intracranial response rate of 91%159. There are ongoing studies under investigation for new selective RET inhibitors165–168.
NTRK fusions
There are several NTRK genes currently identified. These genes serve to encode proteins TRKA, TRKB, and TRKC, which facilitate growth factor receptor binding to induce tyrosine kinase activity, potentiating a cellular signaling cascade168–170. NTRK fusion genes in malignancy were identified back in the 1980s; however, their role in NSCLC specifically is more novel. Current meta-analyses and systematic reviews detail the prevalence to be between 0.10–0.25% in NSCLC patients171–173. NGS is the preferred method for identification of these extremely rare fusion genes174. The first generation NTRK-TKI, arotrectinib is a highly selective ATP competitive inhibitor of tropomyosin receptor kinases (TRK), received FDA approval in 2018 for adult and pediatric patients with solid tumors and NTRK fusion genes without any additional therapy options175,176. Its efficacy was further solidified in a phase I dose escalation study of 70 patients, 8 of whom had TRK gene fusions, and the ORR was 100%177. With overall response rates of 75% in a study including adults and children with 17 unique TRK fusion-positive tumors178, overall adverse events have been reported to be mild. Entrectinib is an oral selective inhibitor of NTRK proteins, ROS 1, and ALK tyrosine kinase, which received FDA approval in 2019 after Marcus et al. proved a DoR rate of 57%, including a complete response of 7% amongst 54 patients with 10 different NTRK fusion-positive malignancies179. There are multiple phase I and II trials under investigation for entrectinib, which include pediatric and adult patients with solid tumors and NTRK fusion genes. 10 patients with NSCLC were included and achieved an ORR of 70%180. To date, there are select ongoing trials comparing targeted therapies for patients with NTRK-positive NSCLC (NCT04302025, NCT04996121, NCT05192642)181–183.
Targets on the horizon
Given the molecularly complex nature of NSCLC and potential targeted therapies, the field is evolving rapidly. There are new targets being identified and studied continuously. There are numerous ongoing studies to combine known targeted therapies as adjunct therapy to other modalities, including chemotherapeutics, ICIs, radiation therapy, and surgical resection. The details of these studies are beyond the scope of this review. For a more complete list of molecular targeted therapies under investigation at the time of this publication, we have included ongoing clinical trials for reference in Table 1.
Table 1. Targetable Mutation therapy, known resistance mechanisms, and ongoing clinical Trials.
* Updated as of 4/28/2023
Conclusions
The discovery of genetic drivers in cancer development, growth, and metastases has individualized cancer treatment. The field continues to evolve rapidly, with numerous ongoing studies examining multiple combinations of therapies and mechanisms of resistance, which opens the door to additional targeted therapies. With this rapidly evolving landscape, special attention must be paid to what therapies have molecular benefits and exploration to ensure these benefits translate to true clinical outcomes for patients.
The peer reviewers who approve this article are:
Balazs Halmos, Department of Oncology, Montefiore/Albert Einstein Cancer Center, Bronx, NY, USA
Apar Kishor Ganti, Division of Oncology-Hematology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
Funding Statement
NCI 5P20CA252717-02 (S.P.N.) supported this manuscript.
The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
References
- 1. Sung H, Ferlay J, Siegel RL, et al. : Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021; 71(3): 209–249. 10.3322/caac.21660 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 2. Cancer of the lung and bronchus - cancer stat facts. Accessed Jan 10, 2022. Reference Source
- 3. Siegel RL, Miller KD, Wagle NS, et al. : Cancer statistics, 2023. CA Cancer J Clin. 2023; 73(1): 17–48. 10.3322/caac.21763 [DOI] [PubMed] [Google Scholar]
- 4. Nicholson AG, Tsao MS, Beasley MB, et al. : The 2021 WHO classification of lung tumors: Impact of advances since 2015. J Thorac Oncol. 2022; 17(3): 362–387. 10.1016/j.jtho.2021.11.003 [DOI] [PubMed] [Google Scholar]
- 5. Basumallik N, Agarwal M: Small cell lung cancer. In: StatPearls. Treasure Island (FL): StatPearls Publishing; 2021; Accessed Jan 3, 2022. Reference Source [PubMed] [Google Scholar]
- 6. Lung cancer survival rates | 5-year survival rates for lung cancer. Accessed Mar 20, 2023. Reference Source
- 7. Herbst RS: Review of epidermal growth factor receptor biology. Int J Radiat Oncol Biol Phys. 2004; 59(2 Suppl): 21–26. 10.1016/j.ijrobp.2003.11.041 [DOI] [PubMed] [Google Scholar]
- 8. Zhang H, Berezov A, Wang Q, et al. : ErbB receptors: From oncogenes to targeted cancer therapies. J Clin Invest. 2007; 117(8): 2051–2058. 10.1172/JCI32278 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Wu Y, Tsuboi M, He J, et al. : Osimertinib in resected EGFR-mutated Non–Small-cell lung cancer. N Engl J Med. 2020; 383(18): 1711–1723. 10.1056/NEJMoa2027071 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 10. Chu QS: Targeting non-small cell lung cancer: Driver mutation beyond epidermal growth factor mutation and anaplastic lymphoma kinase fusion. Ther Adv Med Oncol. 2020; 12: 1758835919895756. 10.1177/1758835919895756 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Kris MG, Johnson BE, Berry LD, et al. : Using multiplexed assays of oncogenic drivers in lung cancers to select targeted drugs. JAMA. 2014; 311(19): 1998–2006. 10.1001/jama.2014.3741 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Mok TS, Wu Y, Thongprasert S, et al. : Gefitinib or Carboplatin–Paclitaxel in pulmonary adenocarcinoma. N Engl J Med. 2009; 361(10): 947–957. 10.1056/NEJMoa0810699 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 13. Mitsudomi T, Morita S, Yatabe Y, et al. : Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor (WJTOG3405): An open label, randomised phase 3 trial. Lancet Oncol. 2010; 11(2): 121–128. 10.1016/S1470-2045(09)70364-X [DOI] [PubMed] [Google Scholar]
- 14. Maemondo M, Inoue A, Kobayashi K, et al. : Gefitinib or chemotherapy for Non–Small-cell lung cancer with mutated EGFR. N Engl J Med. 2010; 362(25): 2380–2388. 10.1056/NEJMoa0909530 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 15. Zhou C, Wu YL, Chen G, et al. : Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer (OPTIMAL, CTONG-0802): A multicentre, open-label, randomised, phase 3 study. Lancet Oncol. 2011; 12(8): 735–742. 10.1016/S1470-2045(11)70184-X [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 16. 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(3): 239–246. 10.1016/S1470-2045(11)70393-X [DOI] [PubMed] [Google Scholar]
- 17. Sequist LV, Yang JCH, 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(27): 3327–3334. 10.1200/JCO.2012.44.2806 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 18. Wu YL, Zhou C, Hu CP, et al. : Afatinib versus cisplatin plus gemcitabine for first-line treatment of asian patients with advanced non-small-cell lung cancer harbouring EGFR mutations (LUX-lung 6): An open-label, randomised phase 3 trial. Lancet Oncol. 2014; 15(2): 213–222. 10.1016/S1470-2045(13)70604-1 [DOI] [PubMed] [Google Scholar]
- 19. Pao W, Miller VA, Politi KA, et al. : Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Med. 2005; 2(3): e73. 10.1371/journal.pmed.0020073 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Mok TS, Wu YL, Ahn MJ, et al. : Osimertinib or Platinum–Pemetrexed in EGFR T790M–Positive lung cancer. N Engl J Med. 2017; 376(7): 629–640. 10.1056/NEJMoa1612674 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 21. Soria JC, Ohe Y, Vansteenkiste J, et al. : Osimertinib in untreated EGFR-mutated advanced Non–Small-cell lung cancer. N Engl J Med. 2018; 378(2): 113–125. 10.1056/NEJMoa1713137 [DOI] [PubMed] [Google Scholar]
- 22. Jänne PA, Yang JCH, Kim DW, et al. : AZD9291 in EGFR Inhibitor–Resistant Non–Small-cell lung cancer. N Engl J Med. 2015; 372(18): 1689–1699. 10.1056/NEJMoa1411817 [DOI] [PubMed] [Google Scholar]
- 23. Yu X, Sheng J, Pan G, et al. : Real‐world utilization of EGFR TKIs and prognostic factors for survival in EGFR‐mutated non‐small cell lung cancer patients with brain metastases. Int J Cancer. 2021; 149(5): 1121–1128. 10.1002/ijc.33677 [DOI] [PubMed] [Google Scholar]
- 24. Ramalingam SS, Vansteenkiste J, Planchard D, et al. : Overall survival with osimertinib in untreated, EGFR-mutated advanced NSCLC. N Engl J Med. 2020; 382(1): 41–50. 10.1056/NEJMoa1913662 [DOI] [PubMed] [Google Scholar]
- 25. 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(8): 2240–2247. 10.1158/1078-0432.CCR-12-2246 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 26. Papadimitrakopoulou VA, Wu YL, Han JY, et al. : LBA51Analysis of resistance mechanisms to osimertinib in patients with EGFR T790M advanced NSCLC from the AURA3 study. Ann Oncol. 2018; 29(suppl_8): viii741. 10.1093/annonc/mdy424.064 [DOI] [Google Scholar]
- 27. Jeong SY, Yun J, Yang SD, et al. : Abstract 1106: BRAF and EGFR fusion as a novel mechanism of resistance mechanism to lazertinib, 3rd- generation EGFR-TKI, in EGFR-mutant NSCLC. Cancer Res. 2021; 81(13_Supplement): 1106. 10.1158/1538-7445.AM2021-1106 [DOI] [Google Scholar]
- 28. Arulananda S, Do H, Musafer A, et al. : Combination osimertinib and gefitinib in C797S and T790M EGFR-mutated non-small cell lung cancer. J Thorac Oncol. 2017; 12(11): 1728–1732. 10.1016/j.jtho.2017.08.006 [DOI] [PubMed] [Google Scholar]
- 29. Niederst MJ, Hu H, Mulvey HE, et al. : The allelic context of the C797S mutation acquired upon treatment with third-generation EGFR inhibitors impacts sensitivity to subsequent treatment strategies. Clin Cancer Res. 2015; 21(17): 3924–3933. 10.1158/1078-0432.CCR-15-0560 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Wang Z, Yang JJ, Huang J, et al. : Lung adenocarcinoma harboring EGFR T790M and in trans C797S responds to combination therapy of first- and third-generation EGFR TKIs and shifts allelic configuration at resistance. J Thorac Oncol. 2017; 12(11): 1723–1727. 10.1016/j.jtho.2017.06.017 [DOI] [PubMed] [Google Scholar]
- 31. Ramalingam SS, Cheng Y, Zhou C, et al. : LBA50Mechanisms of acquired resistance to first-line osimertinib: Preliminary data from the phase III FLAURA study. Ann Oncol. 2018; 29(SUPPLEMENT 8): viii740. 10.1093/annonc/mdy424.063 [DOI] [Google Scholar]
- 32. Uchibori K, Inase N, Araki M, et al. : Brigatinib combined with anti-EGFR antibody overcomes osimertinib resistance in EGFR-mutated non-small-cell lung cancer. Nat Commun. 2017; 8(1): 14768. 10.1038/ncomms14768 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Yang Z, Yang N, Ou Q, et al. : Investigating novel resistance mechanisms to third-generation EGFR tyrosine kinase inhibitor osimertinib in non-small cell lung cancer patients. Clin Cancer Res. 2018; 24(13): 3097–3107. 10.1158/1078-0432.CCR-17-2310 [DOI] [PubMed] [Google Scholar]
- 34. Le X, Puri S, Negrao MV, et al. : Landscape of EGFR-Dependent and -Independent Resistance Mechanisms to Osimertinib and Continuation Therapy Beyond Progression in EGFR-Mutant NSCLC. Clin Cancer Res. 2018; 24(24): 6195–6203. 10.1158/1078-0432.CCR-18-1542 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Sehgal K, Rangachari D, VanderLaan PA, et al. : Clinical benefit of tyrosine kinase inhibitors in advanced lung cancer with EGFR-G719A and other uncommon EGFR mutations. Oncologist. 2021; 26(4): 281–287. 10.1002/onco.13537 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Leventakos K, Kipp BR, Rumilla KM, et al. : S768I mutation in EGFR in patients with lung cancer. J Thorac Oncol. 2016; 11(10): 1798–1801. 10.1016/j.jtho.2016.05.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Yang JCH, Sequist LV, Geater SL, et al. : Clinical activity of afatinib in patients with advanced non-small-cell lung cancer harbouring uncommon EGFR mutations: A combined post-hoc analysis of LUX-lung 2, LUX-lung 3, and LUX-lung 6. Lancet Oncol. 2015; 16(7): 830–838. 10.1016/S1470-2045(15)00026-1 [DOI] [PubMed] [Google Scholar]
- 38. Yang JCH, Schuler M, Popat S, et al. : Afatinib for the treatment of non-small cell lung cancer harboring uncommon EGFR mutations: An updated database of 1023 cases brief report. Front Oncol. 2022; 12: 834704. 10.3389/fonc.2022.834704 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Heigener DF, Schumann C, Sebastian M, et al. : Afatinib in Non-Small cell lung cancer harboring uncommon EGFR mutations pretreated with reversible EGFR inhibitors. Oncologist. 2015; 20(10): 1167–1174. 10.1634/theoncologist.2015-0073 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Cho JH, Lim SH, An HJ, et al. : Osimertinib for patients with non-small-cell lung cancer harboring uncommon EGFR mutations: A multicenter, open-label, phase II trial (KCSG-LU15-09). J Clin Oncol. 2020; 38(5): 488–495. 10.1200/JCO.19.00931 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Bar J, Peled N, Schokrpur S, et al. : Uncommon EGFR mutations on osimertinib, real-life data (UNICORN study): Updated results, brain efficacy, and resistance mechanisms. J Clin Oncol. 2022; 40(16): 9109. 10.1200/JCO.2022.40.16_suppl.9109 [DOI] [Google Scholar]
- 42. Arcila ME, Nafa K, Chaft JE, et al. : EGFR exon 20 insertion mutations in lung adenocarcinomas: Prevalence, molecular heterogeneity, and clinicopathologic characteristics. Mol Cancer Ther. 2013; 12(2): 220–229. 10.1158/1535-7163.MCT-12-0620 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Meador CB, Sequist LV, Piotrowska Z: Targeting EGFR Exon 20 Insertions in Non-Small Cell Lung Cancer: Recent Advances and Clinical Updates. Cancer Discov. 2021; 11(9): 2145–2157. 10.1158/2159-8290.CD-21-0226 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Robichaux JP, Elamin YY, Tan Z, et al. : Mechanisms and clinical activity of an EGFR and HER2 exon 20-selective kinase inhibitor in non-small cell lung cancer. Nat Med. 2018; 24(5): 638–646. 10.1038/s41591-018-0007-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Le X, Cornelissen R, Garassino M, et al. : Poziotinib in non-small-cell lung cancer harboring HER2 exon 20 insertion mutations after prior therapies: ZENITH20-2 trial. J Clin Oncol. 2022; 40(7): 710–718. 10.1200/JCO.21.01323 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Heymach J, Negrao M, Robichaux J, et al. : OA02.06 A phase II trial of poziotinib in EGFR and HER2 exon 20 mutant non-small cell lung cancer (NSCLC). J Thorac Oncol. 2018; 13(10): S323–S324. 10.1016/j.jtho.2018.08.243 [DOI] [Google Scholar]
- 47. Elamin YY, Robichaux JP, Carter BW, et al. : Poziotinib for EGFR exon 20-mutant NSCLC: Clinical efficacy, resistance mechanisms, and impact of insertion location on drug sensitivity. Cancer Cell. 2022; 40(7): 754–767.e6. 10.1016/j.ccell.2022.06.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Riely GJ, Neal JW, Camidge DR, et al. : Activity and safety of mobocertinib (TAK-788) in previously treated Non–Small cell lung cancer with EGFR exon 20 insertion mutations from a phase I/II trial. Cancer Discov. 2021; 11(7): 1688–1699. 10.1158/2159-8290.CD-20-1598 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Zhou C, Ramalingam S, Li B, et al. : OA04.03 mobocertinib in NSCLC with EGFR exon 20 insertions: Results from EXCLAIM and pooled platinum-pretreated patient populations. J Thorac Oncol. 2021; 16(3): S108. 10.1016/j.jtho.2021.01.283 [DOI] [Google Scholar]
- 50. Zhou C, Ramalingam SS, Kim TM, et al. : Treatment outcomes and safety of mobocertinib in platinum-pretreated patients with EGFR exon 20 Insertion–Positive metastatic Non–Small cell lung cancer: A phase 1/2 open-label nonrandomized clinical trial. JAMA Oncol. 2021; 7(12): e214761. 10.1001/jamaoncol.2021.4761 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Yun J, Lee S, Kim S, et al. : Antitumor activity of amivantamab (JNJ-61186372), an EGFR–MET bispecific antibody, in diverse models of EGFR exon 20 Insertion–Driven NSCLC. Cancer Discov. 2020; 10(8): 1194–1209. 10.1158/2159-8290.CD-20-0116 [DOI] [PubMed] [Google Scholar]
- 52. Park K, Haura EB, Leighl NB, et al. : Amivantamab in EGFR exon 20 insertion-mutated non-small-cell lung cancer progressing on platinum chemotherapy: Initial results from the CHRYSALIS phase I study. J Clin Oncol. 2021; 39(30): 3391–3402. 10.1200/JCO.21.00662 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 53. A phase 3, randomized study of amivantamab and lazertinib combination therapy versus osimertinib versus lazertinib as first-line treatment in patients with EGFR-mutated locally advanced or metastatic non-small cell lung cancer. 2022.
- 54. A randomized, open-label phase 3 study of combination amivantamab and carboplatin-pemetrexed therapy, compared with carboplatin-pemetrexed, in patients with EGFR exon 20ins mutated locally advanced or metastatic non-small cell lung cancer. 2022; Accessed Nov 4, 2022. Reference Source
- 55. An open-label phase 1/1b study to evaluate the safety and pharmacokinetics of JNJ-73841937 (lazertinib), a third generation EGFR-TKI, as monotherapy or in combinations with JNJ-61186372, a human bispecific EGFR and cMet antibody in participants with advanced non-small cell lung cancer. 2022; Accessed Nov 4, 2022. Reference Source
- 56. Cadranel J, Ruppert A, Beau-Faller M, et al. : Therapeutic strategy for advanced EGFR mutant non-small-cell lung carcinoma. Crit Rev Oncol Hematol. 2013; 88(3): 477–493. 10.1016/j.critrevonc.2013.06.009 [DOI] [PubMed] [Google Scholar]
- 57. Coleman N, Hong L, Zhang J, et al. : Beyond epidermal growth factor receptor: MET amplification as a general resistance driver to targeted therapy in oncogene-driven non-small-cell lung cancer. ESMO Open. 2021; 6(6): 100319. 10.1016/j.esmoop.2021.100319 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Wang W, Wang H, Lu P, et al. : Crizotinib with or without an EGFR-TKI in treating EGFR-mutant NSCLC patients with acquired MET amplification after failure of EGFR-TKI therapy: A multicenter retrospective study. J Transl Med. 2019; 17(1): 52. 10.1186/s12967-019-1803-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Liam CK, Ahmad AR, Hsia T, et al. : Abstract CT538: Tepotinib + gefitinib in patients with EGFR -mutant NSCLC with MET amplification: Final analysis of INSIGHT. Cancer Res. 2022; 82(12_Supplement): CT538. 10.1158/1538-7445.AM2022-CT538 [DOI] [Google Scholar]
- 60. Shi P, Oh YT, Zhang G, et al. : Met gene amplification and protein hyperactivation is a mechanism of resistance to both first and third generation EGFR inhibitors in lung cancer treatment. Cancer Lett. 2016; 380(2): 494–504. 10.1016/j.canlet.2016.07.021 [DOI] [PubMed] [Google Scholar]
- 61. Solassol J, Vendrell JA, Senal R, et al. : Challenging BRAF/EGFR co-inhibition in NSCLC using sequential liquid biopsies. Lung Cancer. 2019; 133: 45–47. 10.1016/j.lungcan.2019.05.003 [DOI] [PubMed] [Google Scholar]
- 62. Ho C, Liao W, Lin C, et al. : Acquired BRAF V600E mutation as resistant mechanism after treatment with osimertinib. J Thorac Oncol. 2017; 12(3): 567–572. 10.1016/j.jtho.2016.11.2231 [DOI] [PubMed] [Google Scholar]
- 63. Lin C, Shih J, Yu C, et al. : Outcomes in patients with non-small-cell lung cancer and acquired Thr790Met mutation treated with osimertinib: A genomic study. Lancet Respir Med. 2018; 6(2): 107–116. 10.1016/S2213-2600(17)30480-0 [DOI] [PubMed] [Google Scholar]
- 64. Jebbink M, Langen AJD, Monkhorst K, et al. : MA02.07 T-DM1 and osimertinib (TRAEMOS) to target HER2 bypass track resistance in EGFRm+ NSCLC: Interim analysis of a phase II trial. J Thorac Oncol. 2021; 16(10): S891–S892. 10.1016/j.jtho.2021.08.116 [DOI] [Google Scholar]
- 65. Bordi P, Del Re M, Minari R, et al. : From the beginning to resistance: Study of plasma monitoring and resistance mechanisms in a cohort of patients treated with osimertinib for advanced T790M-positive NSCLC. Lung Cancer. 2019; 131: 78–85. 10.1016/j.lungcan.2019.03.017 [DOI] [PubMed] [Google Scholar]
- 66. Urbanska E, Soerensen J, Melchior L, et al. : P76.16 EGFR-L858R NSCLC with pleiotropic resistance mechanisms: T790M, C797S, SCLC-transformation and KRAS, TP53, and BRAF mutations. J Thorac Oncol. 2021; 16(3): S592–S593. 10.1016/j.jtho.2021.01.1073 [DOI] [Google Scholar]
- 67. Levin PA, Mayer M, Hoskin S, et al. : Histologic transformation from adenocarcinoma to squamous cell carcinoma as a mechanism of resistance to EGFR inhibition. J Thorac Oncol. 2015; 10(9): e86–e88. 10.1097/JTO.0000000000000571 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Oh IJ, Hur JY, Park CK, et al. : Clinical activity of pan-HER inhibitors against HER2-mutant lung adenocarcinoma. Clin Lung Cancer. 2018; 19(5): e775–e781. 10.1016/j.cllc.2018.05.018 [DOI] [PubMed] [Google Scholar]
- 69. Garrido-Castro A, Felip E: HER2 driven non-small cell lung cancer (NSCLC): Potential therapeutic approaches. Transl Lung Cancer Res. 2013; 2(2): 122–127. 10.3978/j.issn.2218-6751.2013.02.02 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Mar N, Vredenburgh JJ, Wasser JS: Targeting HER2 in the treatment of non-small cell lung cancer. Lung Cancer. 2015; 87(3): 220–225. 10.1016/j.lungcan.2014.12.018 [DOI] [PubMed] [Google Scholar]
- 71. Kuyama S, Hotta K, Tabata M, et al. : Impact of HER2 gene and protein status on the treatment outcome of cisplatin-based chemoradiotherapy for locally advanced non-small cell lung cancer. J Thorac Oncol. 2008; 3(5): 477–482. 10.1097/JTO.0b013e31816e2ea3 [DOI] [PubMed] [Google Scholar]
- 72. Peters S, Stahel R, Bubendorf L, et al. : Trastuzumab emtansine (T-DM1) in patients with previously treated HER2-overexpressing metastatic Non–Small cell lung cancer: Efficacy, safety, and biomarkers. Clin Cancer Res. 2019; 25(1): 64–72. 10.1158/1078-0432.CCR-18-1590 [DOI] [PubMed] [Google Scholar]
- 73. Smit EF, Nakagawa K, Nagasaka M, et al. : Trastuzumab deruxtecan (T-dxd; DS-8201) in patients with HER-2 mutated metastatic non small cell lung cancer (NSCLC): Interim results of DESTINY-Lung01. J Clin Oncol. 2020; 38(15_suppl): 9504–9504. 10.1200/JCO.2020.38.15_suppl.9504 [DOI] [Google Scholar]
- 74. Jänne PA, Baik C, Su W, et al. : Efficacy and safety of patritumab deruxtecan (HER3-DXd) in EGFR inhibitor-resistant, EGFR-mutated non-small cell lung cancer. Cancer Discov. 2022; 12(1): 74–89. 10.1158/2159-8290.CD-21-0715 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Dziadziuszko R, Smit EF, Dafni U, et al. : Afatinib in NSCLC with HER2 mutations: Results of the prospective, open-label phase II NICHE trial of european thoracic oncology platform (ETOP). J Thorac Oncol. 2019; 14(6): 1086–1094. 10.1016/j.jtho.2019.02.017 [DOI] [PubMed] [Google Scholar]
- 76. Li BT, Smit EF, Goto Y, et al. : Trastuzumab deruxtecan in HER2-mutant Non–Small-cell lung cancer. N Engl J Med. 2022; 386(3): 241–251. 10.1056/NEJMoa2112431 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 77. Goto K, Sang-We K, Kubo T, et al. : LBA55 trastuzumab deruxtecan (T-DXd) in patients (pts) with HER2-mutant metastatic non-small cell lung cancer (NSCLC): Interim results from the phase 2 DESTINY-Lung02 trial. Ann Oncol. 2022; 33(SUPPLEMENT 7): S1422. 10.1016/j.annonc.2022.08.057 [DOI] [Google Scholar]
- 78. Okura N, Nishioka N, Yamada T, et al. : ONO-7475, a novel AXL inhibitor, suppresses the adaptive resistance to initial EGFR-TKI treatment in EGFR-mutated non-small cell lung cancer. Clin Cancer Res. 2020; 26(9): 2244–2256. 10.1158/1078-0432.CCR-19-2321 [DOI] [PubMed] [Google Scholar]
- 79. Hirano T, Yasuda H, Hamamoto J, et al. : Pharmacological and structural characterizations of naquotinib, a novel third-generation EGFR tyrosine kinase inhibitor, in EGFR-mutated non-small cell lung cancer. Mol Cancer Ther. 2018; 17(4): 740–750. 10.1158/1535-7163.MCT-17-1033 [DOI] [PubMed] [Google Scholar]
- 80. Sequist LV, Han JY, Ahn MJ, et al. : Osimertinib plus savolitinib in patients with EGFR mutation-positive, MET-amplified, non-small-cell lung cancer after progression on EGFR tyrosine kinase inhibitors: Interim results from a multicentre, open-label, phase 1b study. Lancet Oncol. 2020; 21(3): 373–386. 10.1016/S1470-2045(19)30785-5 [DOI] [PubMed] [Google Scholar]
- 81. Randomized, placebo-controlled, double-blind phase 1b/2 study of U3-1287 (AMG 888) in combination with erlotinib in EGFR treatment naïve subjects with advanced non-small cell lung cancer (NSCLC) who have progressed on at least one prior chemotherapy. 2021; Accessed Jan 9, 2022. Reference Source
- 82. Sholl LM, Aisner DL, Varella-Garcia M, et al. : Multi-institutional oncogenic driver mutation analysis in lung adenocarcinoma: The lung cancer mutation consortium experience. J Thorac Oncol. 2015; 10(5): 768–777. 10.1097/JTO.0000000000000516 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Rohatgi A, Govindan R: Targeting KRAS G12C mutation in lung adenocarcinoma. Lung Cancer. 2021; 165: 28–33. 10.1016/j.lungcan.2021.12.021 [DOI] [PubMed] [Google Scholar]
- 84. Graziano SL, Poiesz BJ, Newman N, et al. : 568 prognostic significance of K-ras codon 12 mutations and p53 expression in patients with resected stage I and II non-small cell lung cancer (NSCLC). Lung Cancer. 1997; 18(Supplement 1): 146. 10.1016/S0169-5002(97)89948-5 [DOI] [PubMed] [Google Scholar]
- 85. Jänne PA, Rybkin II, Spira AI, et al. : KRYSTAL-1: Activity and safety of adagrasib (MRTX849) in advanced/ metastatic Non–Small-cell lung cancer (NSCLC) harboring KRAS G12C mutation. Eur J Cancer (1990). 2020; 138: S1–S2. 10.1016/S0959-8049(20)31076-5 [DOI] [Google Scholar]
- 86. Sabari JK, Velcheti V, Shimizu K, et al. : Activity of adagrasib (MRTX849) in brain metastases: Preclinical models and clinical data from patients with KRASG12C-mutant non-small cell lung cancer. Clin Cancer Res. 2022; 28(15): 3318–3328. 10.1158/1078-0432.CCR-22-0383 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Research, Center for Drug Evaluation and: FDA grants accelerated approval to sotorasib for KRAS G12C mutated NSCLC. FDA, 2021; Accessed Jan 22, 2022. Reference Source
- 88. The LO: Undruggable KRAS—time to rebrand? Lancet Oncol. 2021; 22(3): 289. 10.1016/S1470-2045(21)00091-7 [DOI] [PubMed] [Google Scholar]
- 89. Hong DS, Fakih MG, Strickler JH, et al. : KRASG12C inhibition with sotorasib in advanced solid tumors. N Engl J Med. 2020; 383(13): 1207–1217. 10.1056/NEJMoa1917239 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Awad MM, Liu S, Rybkin II, et al. : Acquired resistance to KRASG12C inhibition in cancer. N Engl J Med. 2021; 384(25): 2382–2393. 10.1056/NEJMoa2105281 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Passaro A, Peters S: Setting the Benchmark for KRASG12C-Mutated NSCLC. N Engl J Med. 2022; 387(2): 180–183. 10.1056/NEJMe2207902 [DOI] [PubMed] [Google Scholar]
- 92. Misale S, Fatherree JP, Cortez E, et al. : KRAS G12C NSCLC models are sensitive to direct targeting of KRAS in combination with PI3K inhibition. Clin Cancer Res. 2019; 25(2): 796–807. 10.1158/1078-0432.CCR-18-0368 [DOI] [PubMed] [Google Scholar]
- 93. Adachi Y, Ito K, Kimura R, et al. : MO18-1 EMT is a cause of both intrinsic and acquired resistance to KRAS G12C inhibitor in KRAS G12C mutant NSCLC. Ann Oncol. 2021; 32: S307. 10.1016/j.annonc.2021.05.607 [DOI] [PubMed] [Google Scholar]
- 94. Suzuki S, Yonesaka K, Teramura T, et al. : KRAS inhibitor resistance in MET-amplified KRASG12C non-small cell lung cancer induced by RAS- and non-RAS-mediated cell signaling mechanisms. Clin Cancer Res. 2021; 27(20): 5697–5707. 10.1158/1078-0432.CCR-21-0856 [DOI] [PubMed] [Google Scholar]
- 95. Adachi Y, Ito K, Hayashi Y, et al. : Epithelial-to-mesenchymal transition is a cause of both intrinsic and acquired resistance to KRAS G12C inhibitor in KRAS G12C-mutant non-small cell lung cancer. Clin Cancer Res. 2020; 26(22): 5962–5973. 10.1158/1078-0432.CCR-20-2077 [DOI] [PubMed] [Google Scholar]
- 96. A first-in-human study of the safety, pharmacokinetics, pharmacodynamics, and preliminary antitumor activity of JNJ-74699157 in participants with advanced solid tumors harboring the KRAS G12C mutation. 2020; Accessed Jan 22, 2022. Reference Source
- 97. A phase ia/ib dose-escalation and dose-expansion study evaluating the safety, pharmacokinetics, and activity of GDC-6036 as a single agent and in combination with other anti-cancer therapies in patients with advanced or metastatic solid tumors with a KRAS G12C mutation. 2021; Accessed Jan 22, 2022. Reference Source
- 98. A phase ib/II open-label, multi-center dose escalation study of JDQ443 in patients with advanced solid tumors harboring the KRAS G12C mutation. 2021; Accessed Jan 22, 2022. Reference Source
- 99. A phase 1/2, open label study to evaluate the safety, tolerability, pharmacokinetics and efficacy of D-1553 in subjects with advanced or metastatic solid tumors with KRasG12C mutation. 2021. Reference Source
- 100. Gort E, Johnson ML, Hwang JJ, et al. : A phase I, open-label, dose-escalation trial of BI 1701963 as monotherapy and in combination with trametinib in patients with KRAS mutated advanced or metastatic solid tumors. JCO. 2020; 38(15_suppl): TPS3651. 10.1200/JCO.2020.38.15_suppl.TPS3651 [DOI] [Google Scholar]
- 101. A phase II study of AMG 510 in participants with previously treated stage IV or recurrent KRAS G12C mutated non-squamous non-small cell lung cancer (ECOG-ACRIN LUNG-MAP SUB-STUDY). 2021; Accessed Nov 2, 2022. Reference Source
- 102. Hillig RC, Sautier B, Schroeder J, et al. : Discovery of potent SOS1 inhibitors that block RAS activation via disruption of the RAS-SOS1 interaction. Proc Natl Acad Sci U S A. 2019; 116(7): 2551–2560. 10.1073/pnas.1812963116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Mainardi S, Mulero-Sánchez A, Prahallad A, et al. : SHP2 is required for growth of KRAS-mutant non-small-cell lung cancer in vivo. Nat Med. 2018; 24(7): 961–967. 10.1038/s41591-018-0023-9 [DOI] [PubMed] [Google Scholar]
- 104. A phase ib, open-label, multi-center study to characterize the safety, tolerability, and preliminary efficacy of TNO155 in combination with spartalizumab or ribociclib in selected malignancies. 2021; Accessed Jan 22, 2022. Reference Source
- 105. A phase 1/2 trial of MRTX849 in combination with TNO155 in patients with advanced solid tumors with KRAS G12C mutation KRYSTAL 2. 2021.
- 106. M.D ID.: A phase IB/II study of lorlatinib combinations in anaplastic lymphoma kinase-rearranged lung cancer. 2021; Accessed Jan 22, 2022. Reference Source
- 107. An open-label, multi-center, phase I, dose finding study of oral TNO155 in adult patients with advanced solid tumors. 2021; Accessed Jan 22, 2022. Reference Source
- 108. A phase ib/II open-label, multi-center dose escalation study of JDQ443 in patients with advanced solid tumors harboring the KRAS G12C mutation. 2022; Accessed Nov 6, 2022. Reference Source
- 109. Drilon A, Clark JW, Weiss J, et al. : Antitumor activity of crizotinib in lung cancers harboring a MET exon 14 alteration. Nat Med. 2020; 26(1): 47–51. 10.1038/s41591-019-0716-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Middleton G, Fletcher P, Popat S, et al. : The national lung matrix trial of personalized therapy in lung cancer. Nature. 2020; 583(7818): 807–812. 10.1038/s41586-020-2481-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Wolf J, Seto T, Han JY, et al. : Capmatinib in MET exon 14–Mutated or MET-amplified Non–Small-cell lung cancer. N Engl J Med. 2020; 383(10): 944–957. 10.1056/NEJMoa2002787 [DOI] [PubMed] [Google Scholar]
- 112. Paik PK, Felip E, Veillon R, et al. : Tepotinib in Non–Small-cell lung cancer with MET exon 14 skipping mutations. N Engl J Med. 2020; 383(10): 931–943. 10.1056/NEJMoa2004407 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Lu S, Fang J, Li X, et al. : Phase II study of savolitinib in patients (pts) with pulmonary sarcomatoid carcinoma (PSC) and other types of non-small cell lung cancer (NSCLC) harboring MET exon 14 skipping mutations (METex14+). J Clin Oncol. 2020; 38(15_suppl): 9519. 10.1200/JCO.2020.38.15_suppl.9519 [DOI] [Google Scholar]
- 114. Research, Center for Drug Evaluation and: FDA grants accelerated approval to capmatinib for metastatic non-small cell lung cancer. FDA, 2021; Accessed Jan 23, 2022. Reference Source
- 115. Research, Center for Drug Evaluation and: FDA grants accelerated approval to tepotinib for metastatic non-small cell lung cancer. FDA, 2021; Accessed Jan 23, 2022. Reference Source
- 116. A phase III, randomized, controlled, open-label, multicenter, global study of capmatinib versus SoC docetaxel chemotherapy in previously treated patients with EGFR wt, ALK negative, locally advanced or metastatic (stage IIIB/IIIC or IV) NSCLC harboring MET exon 14 skipping mutation (METΔex14). 2021; Accessed Jan 25, 2022. Reference Source
- 117. Recondo G, Bahcall M, Spurr LF, et al. : Molecular mechanisms of acquired resistance to MET tyrosine kinase inhibitors in patients with MET exon 14-mutant NSCLC. Clin Cancer Res. 2020; 26(11): 2615–2625. 10.1158/1078-0432.CCR-19-3608 [DOI] [PubMed] [Google Scholar]
- 118. Li J, Wang Q, Ge J, et al. : BRAF V600E mediates crizotinib resistance and responds to dabrafenib and trametinib in a ROS1-Rearranged Non-Small cell lung cancer: A case report. Oncologist. 2021; 26(12): e2115–e2119. 10.1002/onco.13979 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Cardarella S, Ogino A, Nishino M, et al. : Clinical, pathologic, and biologic features associated with BRAF mutations in non-small cell lung cancer. Clin Cancer Res. 2013; 19(16): 4532–4540. 10.1158/1078-0432.CCR-13-0657 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Wan PTC, Garnett MJ, Roe SM, et al. : Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF. Cell. 2004; 116(6): 855–867. 10.1016/S0092-8674(04)00215-6 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 121. Melosky B, Wheatley-Price P, Juergens RA, et al. : The rapidly evolving landscape of novel targeted therapies in advanced non-small cell lung cancer. Lung Cancer. 2021; 160: 136–151. 10.1016/j.lungcan.2021.06.002 [DOI] [PubMed] [Google Scholar]
- 122. O’Leary CG, Andelkovic V, Ladwa R, et al. : Targeting BRAF mutations in non-small cell lung cancer. Transl Lung Cancer Res. 2019; 8(6): 1119–1124. 10.21037/tlcr.2019.10.22 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Chan XY, Singh A, Osman N, et al. : Role played by signalling pathways in overcoming BRAF inhibitor resistance in melanoma. Int J Mol Sci. 2017; 18(7): 1527. 10.3390/ijms18071527 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Lopez-Chavez A, Thomas A, Rajan A, et al. : Molecular profiling and targeted therapy for advanced thoracic malignancies: A biomarker-derived, multiarm, multihistology phase II basket trial. J Clin Oncol. 2015; 33(9): 1000–1007. 10.1200/JCO.2014.58.2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. 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(10): 1307–1316. 10.1016/S1470-2045(17)30679-4 [DOI] [PubMed] [Google Scholar]
- 126. 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(7): 984–993. 10.1016/S1470-2045(16)30146-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Dudnik E, Bar J, Peled N, et al. : Efficacy and safety of BRAF inhibitors with or without MEK inhibitors in BRAF-mutant advanced Non-Small-cell lung cancer: Findings from a real-life cohort. Clin Lung Cancer. 2019; 20(4): 278–286.e1. 10.1016/j.cllc.2019.03.007 [DOI] [PubMed] [Google Scholar]
- 128. Mazieres J, Cropet C, Montané L, et al. : Vemurafenib in non-small-cell lung cancer patients with BRAFV600 and BRAFnonV600 mutations. Ann Oncol. 2020; 31(2): 289–294. 10.1016/j.annonc.2019.10.022 [DOI] [PubMed] [Google Scholar]
- 129. Subbiah V, Gervais R, Riely G, et al. : Efficacy of vemurafenib in patients with non-small-cell lung cancer with BRAF V600 mutation: An open-label, single-arm cohort of the histology-independent VE-BASKET study. JCO Precis Oncol. 2019; 3: 1–9. 10.1200/PO.18.00266 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Research, Center for Drug Evaluation: FDA grants regular approval to dabrafenib and trametinib combination for metastatic NSCLC with BRAF V600E mutation. FDA, 2019; Accessed Jan 23, 2022. Reference Source
- 131. Myall NJ, Henry S, Wood D, et al. : Natural disease history, outcomes, and co-mutations in a series of patients with BRAF-mutated non-small-cell lung cancer. Clin Lung Cancer. 2019; 20(2): e208–e217. 10.1016/j.cllc.2018.10.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Hirai N, Hatanaka Y, Hatanaka KC, et al. : Cyclin-dependent kinase 4 upregulation mediates acquired resistance of dabrafenib plus trametinib in BRAF V600E-mutated lung cancer. Transl Lung Cancer Res. 2021; 10(9): 3737–3744. 10.21037/tlcr-21-415 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. An open label, multicenter phase II clinical trial to evaluate the Efficacy, Safety and pharmacokinetics of HLX208 in advanced non-small cell lung cancer patients with BRAF V600 mutation. 2022.
- 134. A phase II study of the BRAF inhibitor encorafenib in combination with the MEK inhibitor binimetinib in patients with BRAFV600E-mutant metastatic non-small cell lung cancer. 2022.
- 135. A phase 2 study of VS-6766 (dual RAF/MEK inhibitor) as a single agent and in combination with defactinib (FAK inhibitor) in recurrent KRAS-mutant (KRAS-MT) and BRAF-mutant (BRAF-MT) non-small cell lung cancer (NSCLC) (RAMP 202). 2022; Accessed Oct 23, 2022. Reference Source
- 136. Multicenter, open-label, phase II study with a safety lead-in part investigating the efficacy, safety and pharmacokinetics of encorafenib and binimetinib combination in BRAF V600E mutated chinese patients with metastatic non-small cell lung cancer who are BRAF- and MEK inhibitor treatment-naïve. 2022; Accessed Oct 23, 2022. Reference Source
- 137. Choi YL, Soda M, Yamashita Y, et al. : EML4-ALK mutations in lung cancer that confer resistance to ALK inhibitors. N Engl J Med. 2010; 363(18): 1734–1739. 10.1056/NEJMoa1007478 [DOI] [PubMed] [Google Scholar]
- 138. Koivunen JP, Mermel C, Zejnullahu K, et al. : EML4-ALK fusion gene and efficacy of an ALK kinase inhibitor in lung cancer. Clin Cancer Res. 2008; 14(13): 4275–4283. 10.1158/1078-0432.CCR-08-0168 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 139. 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(7153): 561–566. 10.1038/nature05945 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 140. Malik SM, Maher VE, Bijwaard KE, et al. : U.S. food and drug administration approval: Crizotinib for treatment of advanced or metastatic non-small cell lung cancer that is anaplastic lymphoma kinase positive. Clin Cancer Res. 2014; 20(8): 2029–2034. 10.1158/1078-0432.CCR-13-3077 [DOI] [PubMed] [Google Scholar]
- 141. 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(10): e5–e11. 10.1634/theoncologist.2014-0241 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Moro-Sibilot D, Cozic N, Pérol M, et al. : Crizotinib in c-MET- or ROS1-positive NSCLC: results of the AcSé phase II trial. Ann Oncol. 2019; 30(12): 1985–1991. 10.1093/annonc/mdz407 [DOI] [PubMed] [Google Scholar]
- 143. Shaw AT, Ou SI, Bang YJ, et al. : Crizotinib in ROS1-rearranged Non–Small-cell lung cancer. N Engl J Med. 2014; 371(21): 1963–1971. 10.1056/NEJMoa1406766 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Shaw AT, Bauer TM, de Marinis F, et al. : First-line lorlatinib or crizotinib in advanced ALK-positive lung cancer. N Engl J Med. 2020; 383(21): 2018–2029. 10.1056/NEJMoa2027187 [DOI] [PubMed] [Google Scholar]
- 145. 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(23): 2613–2618. 10.1200/JCO.2016.71.3701 [DOI] [PubMed] [Google Scholar]
- 146. Felip E, Bauer T, Solomon B, et al. : MA07.11 safety and efficacy of lorlatinib (PF-06463922) in patients with advanced ALK+ or ROS1+ non-small-cell lung cancer (NSCLC). J Thorac Oncol. 2017; 12(1): S383–S384. 10.1016/j.jtho.2016.11.433 [DOI] [Google Scholar]
- 147. 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(4): 452–463. 10.1016/S1470-2045(15)00614-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Camidge DR, Kim HR, Ahn MJ, et al. : Brigatinib versus crizotinib in ALK Inhibitor-Naive advanced ALK-positive NSCLC: Final results of phase 3 ALTA-1L trial. J Thorac Oncol. 2021; 16(12): 2091–2108. 10.1016/j.jtho.2021.07.035 [DOI] [PubMed] [Google Scholar]
- 149. Tanimoto A, Matsumoto S, Takeuchi S, et al. : Proteasome inhibition overcomes ALK-TKI resistance in ALK-rearranged/ TP53-mutant NSCLC via noxa expression. Clin Cancer Res. 2021; 27(5): 1410–1420. 10.1158/1078-0432.CCR-20-2853 [DOI] [PubMed] [Google Scholar]
- 150. Urbanska EM, Sørensen JB, Melchior LC, et al. : Changing ALK-TKI-resistance mechanisms in rebiopsies of ALK-rearranged NSCLC: ALK- and BRAF-mutations followed by epithelial-mesenchymal transition. Int J Mol Sci. 2020; 21(8): 2847. 10.3390/ijms21082847 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Krebs MG, De Braud F, Siena S, et al. : 1287P efficacy and safety of entrectinib in locally advanced/metastatic ROS1 fusion-positive NSCLC: An updated integrated analysis. Ann Oncol. 2020; 31: S831–S833. 10.1016/j.annonc.2020.08.1601 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Papadopoulos KP, Borazanci E, Shaw AT, et al. : U.S. phase I first-in-human study of taletrectinib (DS-6051b/AB-106), a ROS1/TRK inhibitor, in patients with advanced solid tumors. Clin Cancer Res. 2020; 26(18): 4785–4794. 10.1158/1078-0432.CCR-20-1630 [DOI] [PubMed] [Google Scholar]
- 153. Doebele RC, Lin JJ, Nagasaka M, et al. : TRIDENT-1: A global, multicenter, open-label phase II study investigating the activity of repotrectinib in advanced solid tumors harboring ROS1 or NTRK1-3 rearrangements. J Clin Oncol. 2020; 38(15): TPS9637. 10.1200/JCO.2020.38.15_suppl.TPS9637 [DOI] [Google Scholar]
- 154. De Braud FG, Niger M, Damian S, et al. : Alka-372-001: First-in-human, phase I study of entrectinib – an oral pan-trk, ROS1, and ALK inhibitor – in patients with advanced solid tumors with relevant molecular alterations. JCO. 2015; 33(15_suppl): 2517–2517. 10.1200/jco.2015.33.15_suppl.2517 [DOI] [Google Scholar]
- 155. Patel MR, Bauer TM, Liu SV, et al. : STARTRK-1: Phase 1/2a study of entrectinib, an oral pan-trk, ROS1, and ALK inhibitor, in patients with advanced solid tumors with relevant molecular alterations. JCO. 2015; 33(15_suppl): 2596–2596. 10.1200/jco.2015.33.15_suppl.2596 [DOI] [Google Scholar]
- 156. Mazieres J, Drilon A, Lusque A, et al. : Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: Results from the IMMUNOTARGET registry. Ann Oncol. 2019; 30(8): 1321–1328. 10.1093/annonc/mdz167 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Dantoing E, Piton N, Salaün M, et al. : Anti-PD1/PD-L1 immunotherapy for non-small cell lung cancer with actionable oncogenic driver mutations. Int J Mol Sci. 2021; 22(12): 6288. 10.3390/ijms22126288 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Mushtaq R, Cortot AB, Gautschi O, et al. : PD-1/PD-L1 inhibitor activity in patients with gene-rearrangement positive non-small cell lung cancer—an IMMUNOTARGET case series. Transl Lung Cancer Res. 2022; 11(12): 2412–2417. 10.21037/tlcr-22-329 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Drilon A, Oxnard GR, Tan DSW, et al. : Efficacy of selpercatinib in RET Fusion–Positive Non–Small-cell lung cancer. N Engl J Med. 2020; 383(9): 813–824. 10.1056/NEJMoa2005653 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160. Drilon A, Subbiah V, Gautschi O, et al. : Selpercatinib in patients with RET fusion-positive non-small-cell lung cancer: Updated safety and efficacy from the registrational LIBRETTO-001 phase I/II trial. J Clin Oncol. 2023; 41(2): 385–394. 10.1200/JCO.22.00393 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Subbiah V, Gainor JF, Oxnard GR, et al. : Intracranial efficacy of selpercatinib in RET fusion-positive non-small cell lung cancers on the LIBRETTO-001 trial. Clin Cancer Res. 2021; 27(15): 4160–4167. 10.1158/1078-0432.CCR-21-0800 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. 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(12): 1653–1660. 10.1016/S1470-2045(16)30562-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. 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(1): 42–50. 10.1016/S2213-2600(16)30322-8 [DOI] [PubMed] [Google Scholar]
- 164. Gainor JF, Curigliano G, Kim D, et al. : Registrational dataset from the phase I/II ARROW trial of pralsetinib (BLU-667) in patients (pts) with advanced RET fusion+ non-small cell lung cancer (NSCLC). J Clin Oncol. 2020; 38(15): 9515. 10.1200/JCO.2020.38.15_suppl.9515 [DOI] [Google Scholar]
- 165. A phase 1/2 study of oral selpercatinib (LOXO-292) in patients with advanced solid tumors, including RET fusion-positive solid tumors, medullary thyroid cancer, and other tumors with RET activation (LIBRETTO-001). 2022; Accessed Oct 21, 2022. Reference Source
- 166. Drilon A, Fu S, Patel MR, et al. : A phase I/ib trial of the VEGFR-sparing multikinase RET inhibitor RXDX-105. Cancer Discov. 2019; 9(3): 384–395. 10.1158/2159-8290.CD-18-0839 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. LIBRETTO-431: A multicenter, randomized, open-label, phase 3 trial comparing selpercatinib to platinum-based and pemetrexed therapy with or without pembrolizumab as initial treatment of advanced or metastatic RET fusion-positive non-small cell lung cancer. 2022; Accessed Jan 25, 2022. Reference Source
- 168. A phase III randomized, open-label study of pralsetinib versus standard of care for first-line treatment of RET fusion-positive, metastatic non-small cell lung cancer. 2021. Reference Source
- 169. Lamballe F, Klein R, Barbacid M: trkC, a new member of the trk family of tyrosine protein kinases, is a receptor for neurotrophin-3. Cell. 1991; 66(5): 967–979. 10.1016/0092-8674(91)90442-2 [DOI] [PubMed] [Google Scholar]
- 170. Cordon-Cardo C, Tapley P, Jing SQ, et al. : The trk tyrosine protein kinase mediates the mitogenic properties of nerve growth factor and neurotrophin-3. Cell. 1991; 66(1): 173–183. 10.1016/0092-8674(91)90149-s [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Forsythe A, Zhang W, Phillip Strauss U, et al. : A systematic review and meta-analysis of neurotrophic tyrosine receptor kinase gene fusion frequencies in solid tumors. Ther Adv Med Oncol. 2020; 12: 1758835920975613. 10.1177/1758835920975613 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Gatalica Z, Xiu J, Swensen J, et al. : Molecular characterization of cancers with NTRK gene fusions. Mod Pathol. 2019; 32(1): 147–153. 10.1038/s41379-018-0118-3 [DOI] [PubMed] [Google Scholar]
- 173. Rosen EY, Goldman DA, Hechtman JF, et al. : TRK fusions are enriched in cancers with uncommon histologies and the absence of canonical driver mutations. Clin Cancer Res. 2020; 26(7): 1624–1632. 10.1158/1078-0432.CCR-19-3165 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Farago AF, Taylor MS, Doebele RC, et al. : Clinicopathologic features of non-small-cell lung cancer harboring an NTRK gene fusion. JCO Precis Oncol. 2018; 2018: PO.18.00037. 10.1200/PO.18.00037 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Scott LJ: Larotrectinib: First global approval. Drugs. 2019; 79(2): 201–206. 10.1007/s40265-018-1044-x [DOI] [PubMed] [Google Scholar]
- 176. Hong DS, DuBois SG, Kummar S, et al. : Larotrectinib in patients with TRK fusion-positive solid tumours: A pooled analysis of three phase 1/2 clinical trials. Lancet Oncol. 2020; 21(4): 531–540. 10.1016/S1470-2045(19)30856-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Hong DS, Bauer TM, Lee JJ, et al. : Larotrectinib in adult patients with solid tumours: A multi-centre, open-label, phase I dose-escalation study. Ann Oncol. 2019; 30(2): 325–331. 10.1093/annonc/mdy539 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. 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(8): 731–739. 10.1056/NEJMoa1714448 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 179. Marcus L, Donoghue M, Aungst S, et al. : FDA approval summary: Entrectinib for the treatment of NTRK gene fusion solid tumors. Clin Cancer Res. 2021; 27(4): 928–932. 10.1158/1078-0432.CCR-20-2771 [DOI] [PubMed] [Google Scholar]
- 180. Doebele RC, Drilon A, Paz-Ares L, et al. : Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: Integrated analysis of three phase 1-2 trials. Lancet Oncol. 2020; 21(2): 271–282. 10.1016/S1470-2045(19)30691-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181. A Study of Multiple Therapies in Biomarker-Selected Patients With Resectable Stages IB-III Non-Small Cell Lung Cancer. 2023; Accessed May 29, 2023. Reference Source
- 182. A Study of XZP-5955 Tablets in Patients With NTRK or ROS1 Fusion Positive Locally Advanced or Metastatic Solid Tumors. 2023; Accessed May 29, 2023. Reference Source
- 183. A Study Called VICTORIA to Learn More About How Well Larotrectinib Works in Adults With TRK Fusion-positive Cancer by Comparing Larotrectinib Data From Clinical Studies With Data of Other Treatments From Actual Practice. 2023; Accessed May 29, 2023. Reference Source
