Next Article in Journal
Claudins in Cancer: A Current and Future Therapeutic Target
Previous Article in Journal
The Immune Response of OAS1, IRF9, and IFI6 Genes in the Pathogenesis of COVID-19
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

RAF and MEK Inhibitors in Non-Small Cell Lung Cancer

by
Christos Adamopoulos
1,2,*,
Kostas A. Papavassiliou
3,
Poulikos I. Poulikakos
2 and
Athanasios G. Papavassiliou
1,*
1
Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
2
Department of Oncological Sciences, Precision Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
3
First University Department of Respiratory Medicine, ‘Sotiria’ Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(9), 4633; https://doi.org/10.3390/ijms25094633
Submission received: 29 February 2024 / Revised: 17 April 2024 / Accepted: 22 April 2024 / Published: 24 April 2024
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)

Abstract

:
Lung cancer, despite recent advancements in survival rates, represents a significant global health burden. Non-small cell lung cancer (NSCLC), the most prevalent type, is driven largely by activating mutations in Kirsten rat sarcoma viral oncogene homologue (KRAS) and receptor tyrosine kinases (RTKs), and less in v-RAF murine sarcoma viral oncogene homolog B (BRAF) and mitogen-activated protein-kinase kinase (MEK), all key components of the RTK-RAS-mitogen-activated protein kinase (MAPK) pathway. Learning from melanoma, the identification of BRAFV600E substitution in NSCLC provided the rationale for the investigation of RAF and MEK inhibition as a therapeutic strategy. The regulatory approval of two RAF-MEK inhibitor combinations, dabrafenib–trametinib, in 2017, and encorafenib–binimetinib, in 2023, signifies a breakthrough for the management of BRAFV600E-mutant NSCLC patients. However, the almost universal emergence of acquired resistance limits their clinical benefit. New RAF and MEK inhibitors, with distinct biochemical characteristics, are in preclinical and clinical development. In this review, we aim to provide valuable insights into the current state of RAF and MEK inhibition in the management of NSCLC, fostering a deeper understanding of the potential impact on patient outcomes.

1. Introduction

Lung cancer remains the leading cause of cancer-related mortality worldwide, accounting for 18% of all cancer-related deaths, despite the increased survival rates over the last years [1,2]. In the United States alone, it will account for an estimated 125,070 deaths in the year 2024 [3]. Lung cancer is broadly divided histologically into non-small cell lung cancer (NSCLC), which accounts for 85% of the cases, and small cell lung cancer (SCLC). NSCLC is categorized further into several histological subtypes, of which the most prevalent are lung adenocarcinoma (LUAD), lung squamous cell carcinoma (LSCC), and large cell carcinoma accounting for about 40%, 25%, and 10% of NSCLC cases, respectively [4,5]. The complex molecular landscape in NSCLC comprises genetic alterations in a wide range of genes coding mainly for key and druggable components of the receptor tyrosine kinase (RTK)-RAS-mitogen-activated protein kinase (MAPK) axis. These include Kirsten rat sarcoma (KRAS), epidermal growth factor receptor (EGFR), v-RAF murine sarcoma viral oncogene homolog B (BRAF), anaplastic lymphoma kinase (ALK), c-ROS oncogene-1 (ROS1), hepatocyte growth factor receptor (HGFR or MET), human epidermal growth factor receptor 2 (HER2), rearranged during transfection (RET), and neurotrophic tropomyosin receptor kinase (NTRK) [6,7]. All of these molecular aberrations can drive lung cancer oncogenesis. The implementation of targeted therapies in the clinical practice based on molecular profiling of NSCLC patients has led to a significant improvement in lung cancer survival rates [1,8,9]. The identification of BRAF mutations, particularly the substitution V600E in about 4% of NSCLC, and the clinical success of BRAF and MEK inhibitors in the context of metastatic BRAFV600E-mutant melanoma set the rationale for the clinical investigation of these drugs as a therapeutic strategy targeting BRAFV600E-mutant NSCLC. Over the last years, the Food and Drug Administration (FDA) has granted approval for BRAF and MEK inhibitor combinations for the treatment of BRAFV600E-mutant NSCLC. These include the 2017 approval for the combination of dabrafenib (BRAF inhibitor) with trametinib (MEK inhibitor) and the more recent (2023) approval for the combination of encorafenib (BRAF inhibitor) with binimetinib (MEK inhibitor). Despite these advancements, notable challenges persist whenn utilizing BRAF and MEK inhibitors for the management of BRAF mutant-driven lung cancer. Foremost among these challenges is the emergence of adaptive resistance to BRAF and MEK inhibition, which hinders therapeutic efforts [10,11]. Secondarily, the use of these inhibitors is limited to the case of BRAFV600E-mutant NSCLC, which has a low frequency among lung cancer cases. There is an urgent need for the development of rationally designed combinatorial treatment strategies that can overcome the development of drug resistance, while simultaneously expanding therapeutic indications, to BRAF non-V600-, KRAS-, or RTK-mutant NSCLC. The next generation of RAF and MEK inhibitors, with distinct biochemical properties, are already under preclinical and clinical evaluation for the treatment of lung cancer. This review aims to explore the current state of knowledge and therapeutic advancements of RAF and MEK inhibitors’ evaluation in preclinical and clinical cases of mostly BRAF-mutant lung cancer hoping to provide new insights for a more precise and rational therapeutic design and pharmacological intervention.

2. The RTK/RAS/MAPK Pathway in Lung Cancer

2.1. RTK/RAS/MAPK Pathway

The RTK/RAS/MAPK pathway is a fundamental signal transduction cascade which, in normal cells, conveys extracellular signals from cell surface receptors, mainly RTKs, intracellularly to the nucleus to promote several cellular functions such as survival, cell growth, and differentiation [12,13]. Under normal conditions, stimulation of RTKs, usually upon growth factor binding, results in their intracellular activation and in the recruitment and activation of adaptor proteins such as the Src homology region 2 domain-containing phosphatase 2 (SHP2) and the growth factor receptor-bound protein 2 (GRB2). These adaptor proteins transmit signals that result in the recruitment and activation of guanine nucleotide-exchange factors (GEFs), such as son of sevenless homologue 1 (SOS1). GEFs directly activate the membrane-bound small GTPase RAS, switching it from an inactive GDP-bound form to an active GTP-bound form. Active RAS in turn recruits and activates, through dimerization and phosphorylation, members of the rapidly accelerated fibrosarcoma (RAF) kinase family (ARAF, BRAF, and CRAF) through interaction with their N-terminal RAS-binding domain. The dimerization of two RAF protomers results in the formation of a catalytically active protein dimer [12,14]. The formed catalytically active RAF dimer is stabilized structurally through an inward movement of the αC-helix [15,16]. Activated RAF facilitates phosphorylation and activation of MEK1/2, which can then phosphorylate and activate ERK1/Activated ERK1/2, and can then phosphorylate several downstream targets of the MAPK pathway, such as transcriptions factors, including AP-1, c-MYC, and ELK-1, that control cell survival and proliferation [17,18,19]. At the same time, activated ERK induce multiple negative feedback mechanisms to regulate excessive MAPK pathway activity. These include, among other mechanisms, the immediate suppression of RTKs’ expression upstream of RAF [11,20]. Oncogenic mutations in key components of the RTK/RAS/MAPK pathway drive up to 30% of all human cancers. NSCLC, specifically, is driven by, usually mutually exclusive, mutations in RTKs, such as EGFR (1–15%, in non-Asian populations), HER2 (2–4%), ALK (3–7%), ROS1 (1–4%), KRAS (25%), and BRAF (3–5%) (Figure 1) [6,9,18,21,22,23].

2.2. BRAF Inhibitors

The discovery of BRAF mutations, particularly BRAFV600E mutation, as drivers in various cancers [24] provided a rationale for pharmacological targeting of the BRAF oncoprotein. Therefore, RAF inhibitors were developed aiming to disrupt the aberrant oncogenic signaling arising from mutated BRAF. The first-generation, ATP-competitive RAF inhibitors showed disappointing clinical results, mainly because of poor MAPK pathway inhibition and poor target selectivity [11,25,26]. Next, second-generation, ATP-competitive RAF inhibitors were developed, which have increased selectivity for RAF. These RAF inhibitors, belonging structurally to the Type 1.5 class (αC-helix-OUT, DGF-IN) [27], include vemurafenib, dabrafenib, and encorafenib and have demonstrated impressive clinical activity [28,29] (Figure 1). Notably, this class of RAF inhibitors was effective in suppressing the MAPK pathway only in BRAFV600E-mutant cancer cells and not in BRAF-wild type cells [28,29,30]. This characteristic is responsible for their broad therapeutic window when targeting BRAFV600E tumors. Moreover, inhibitors of this class induce paradoxical activation of MAPK signaling in both BRAF-wild type normal and cancer cells [31,32,33]. The base of this paradox is the differential selectivity of these inhibitors for the monomeric versus dimeric form of BRAF. Thus, type 1.5 RAF inhibitors, herein RAF monomer-selective inhibitors, selectively bind to and inhibit RAF monomers but not dimers. The structural base of this selectivity is that upon binding, the inhibitor stabilizes RAF in a closed inactive conformation, with an outward position of the αC helix (αC-helix OUT RAF inhibitors) [15]. In the case of RAF dimers in cells, these inhibitors bind to one protomer of the dimer while transactivating the unbound protomer (negative allostery) [15]. These discoveries further uncovered the fact that the BRAFV600E oncoprotein signals as a monomer independently of upstream RTK/RAS activation and at the same time sets the base for the classification of BRAF mutations according to their RAF dimerization ability and their dependence on upstream RAS activity [11,13,16,34]. Next-generation equipotent RAF inhibitors, with equal potency for both RAF monomers and dimers, and dimer selective RAF inhibitors, with increased potency for RAF dimers, have been developed [15,35,36,37,38,39,40] (Figure 1). These inhibitors bind to both protomers of the RAF dimer, stabilizing the αC-helix in the IN position (αC-helix IN/DFG-OUT RAF inhibitors or type 2) [15], and are currently under preclinical and clinical evaluation in solid BRAF-mutant cancers, including lung cancer (Table 1).

2.3. Classification of BRAF Mutations

Class I BRAF mutations exclusively include the BRAFV600 substitutions that produce constitutively activated BRAF monomers [34,41]. Class II BRAF mutations form constitutively active RAF dimers with intermediate to high kinase activity that signal independently of RAS, including certain non-V600 missense mutations, splice variants, in-frame deletions, and BRAF fusions [34,41]. Class III BRAF mutations are low-activity, kinase-impaired or kinase-dead mutants that form RAF heterodimers with wild-type CRAF and depend on RAS activity. Often, class III BRAF mutants in tumors co-exist with RAS mutations or neurofibromin 1 (NF1) deletions (melanomas) or RTK upregulation (lung and colorectal cancers), highlighting the need for concurrent mechanisms for sustaining RAS activation despite ERK-dependent feedback [34].

2.4. BRAF Alterations in NSCLC

BRAFV600E mutations are found in about 2% of BRAF-mutant NSCLC patients [7,42,43]. BRAFV600E mutations in NSCLC tend to be more prevalent in cases characterized by micropapillary patterns and among female individuals with a history of smoking. Conversely, BRAF non-V600E mutations are more frequently associated with mucinous patterns and male individuals with a smoking history [42]. A retrospective analysis of BRAF-mutant NSCLC patients demonstrated an association of BRAF mutations, more significantly class II and III compared to class I, with brain metastasis in 29% of patients [44]. In a molecular characterization study of BRAF-mutant NSCLC, the most common BRAF mutations were missense mutations (90%), with half of them being variants of unknown significance [43]. Class I mutations were exclusively BRAFV600E, while G469A and K601E and G466V and N581S were the most common class II and III BRAF mutations, respectively [43]. The same analysis revealed that the most common concurrent mutations were TP53, EGFR, KRAS, and NF1 mutations and that all three classes of BRAF mutations co-existed with the EGFRL858R activating mutation or EGFR exon 19 deletions in 10% of samples [43]. When the association between KRAS and BRAF mutations was examined, it was observed that class III BRAF mutations are more likely to co-occur with KRAS mutations compared to class I and II mutations [43]. Moreover, in another study, BRAF-mutant NSCLC was characterized by elevated levels of PD-L1 expression, in a reported 42% of BRAFV600E and 50% of non-V600E mutations [45]. Conversely, BRAF fusions were rarely detected in 0.2% of a total of 17,128 NSCLC samples, with the most prevalent fusion partners including acylglycerol kinase (AGK), dedicator of cytokinesis protein 4 (DOCK4), and tripartite motif-containing 24 (TRIM24), while the most frequently co-occurring mutations were TP53, CDKN2A, EGFR, and CDKN2B [46]. The prognostic value of BRAFV600E in NSCLC, given its low frequency, is still unclear. However, it has been associated with patients with poor outcomes and low response rates to platinum-based chemotherapy [47].

2.5. MEK Inhibitors

As of now, the FDA has granted approval to four MEK inhibitors: trametinib, selumetinib, cobimetinib, and binimetinib. These small molecule inhibitors are allosteric non-competitive inhibitors of MEK and were the first selective inhibitors of the MAPK pathway to enter the clinic [48]. Trametinib, as a single agent, was approved in 2013 by the FDA for the treatment of metastatic BRAFV600E/K-mutant melanoma patients who have not been previously treated with BRAF inhibitors [49]. Additionally, trametinib is approved in combination with dabrafenib for metastatic patients with BRAFV600E/K-mutant melanoma and BRAFV600E-mutant NSCLC [50], and pediatric patients with BRAFV600E-mutant low-grade glioma with a BRAFV600E mutation. Selumetinib was granted approval for pediatric patients (2 years old and older) with neurofibromatosis type 1 (NF1) who present symptomatic, inoperable plexiform neurofibromas [51]. Cobimetinib is approved in combination with vemurafenib for unresectable or metastatic BRAFV600E/K-mutant melanoma or as a single agent for histiocytic neoplasms [52], while binimetinib in combination with encorafenib is approved for unresectable or metastatic BRAFV600E/K-mutant melanoma and metastatic BRAFV600E-mutant NSCLC [53]. Biochemically, allosteric MEK inhibitors can be divided into two main groups. The first includes compounds that their binding to MEK promotes, with different potencies, the disruption of the RAF−MEK complex, such as trametinib and selumetinib, and a second group which comprises inhibitors that foster MEK−RAF complex formation “RAF/MEK clamps” while preventing phosphorylation by RAF, such as avutometinib [40,54,55,56]. Moreover, although the majority of MEK inhibitors bind to MEK in a similar fashion, occupying the same allosteric pocket, the increased binding affinity of certain MEK inhibitors is the basis for their higher inhibitory potency [40].

2.6. Classification of MEK Mutations

MEK mutations are classified into three distinct types: RAF-independent, RAF-regulated, and RAF-dependent. RAF-independent MEK mutations are typically marked by in-frame deletions resulting in MEK hyperactivation. Conversely, RAF-regulated and RAF-dependent MEK mutations require RAF phosphorylation for their optimal activity [41,57].

2.7. MEK Alterations in NSCLC

The landscape of MEK alterations in lung cancer has remained relatively unexplored due to their extremely rare prevalence. However, when they are present in NSCLC, they are mutually exclusive with other genetic alterations [58,59]. The first study which reported MEK alterations in 2 out of 207 NSCLC patients, identified MAP2K1-K57N mutations and demonstrated their gain-of-function properties in vitro [58]. This was confirmed in a study which reported a 0.6% occurrence of MEK mutations among 6024 lung adenocarcinoma cases, revealed their mutual exclusivity with other oncogenic drivers, and demonstrated an apparent association with current smoking status [59].

3. BRAF and MEK Inhibitors in Lung Cancer

3.1. BRAF Inhibitors as Monotherapy in NSCLC

Since the development of the first RAF monomer-selective inhibitor, vemurafenib, and its successful implementation for the treatment of metastatic BRAFV600E-melanoma, these inhibitors have been evaluated in other tumor types bearing the same mutation [29,60,61]. In the context of BRAFV600E-mutant NSCLC, initial case reports showed a favorable response after vemurafenib administration [62,63]. The effect of vemurafenib, mainly, but also dabrafenib, was evaluated in a retrospective multicenter study in advanced BRAF-mutant NSCLC patients. Vemurafenib treatment showed improved outcomes, with a 54% response rate and 96% disease control, in BRAFV600E patients, while non-V600E patients had poorer outcomes [64]. These results confirm the activity of RAF monomer-selective inhibitors in patients with BRAF-mutant lung cancers. In a phase 2 basket trial, vemurafenib showed promise in BRAFV600-mutation-positive non-melanoma cancers, including NSCLC. Among NSCLC patients, 42% achieved an objective response, with a 23% progression-free survival (PFS) at 12 months. In an expanded cohort, previously untreated patients had a 37.5% objective response rate (ORR), while that of pretreated patients was 37.0%. The median duration of response (DOR) was 7.2 months. Overall, median PFS and median overall survival (OS) were 6.5 and 15.4 months, respectively [65]. Moreover, during the evaluation of vemurafenib in a phase II clinical trial, involving NSCLC patients with both BRAFV600 and non-V600 mutations, no objective responses were observed in the non-V600 cohort, indicating a lack of vemurafenib activity in tumors expressing these BRAF alterations. In stark contrast, the BRAFV600E-cohort demonstrated a remarkable ORR of 44.8%, a 6.4-month median DOR and a 10-month overall survival [66]. Furthermore, there is some retrospective clinical evidence for vemurafenib activity in metastatic lung cancer cases, including brain and leptomeningeal metastases [67,68]. Regarding dabrafenib activity in BRAFV600E-mutant NSCLC, a favorable response was reported in a patient who had been enrolled in a trial for BRAF-mutant melanoma patients [69]. An open-label, multicenter phase II trial evaluating dabrafenib in patients with advanced BRAFV600E-mutant NSCLC showed improved outcomes for both treatment-naïve patients and those chemotherapy pretreated [47]. Additionally, a novel RAF dimer selective inhibitor, lifirafenib (BGB-283), tested in a phase I clinical study, demonstrated promising antitumor activity in KRASG12-mutant NSCLC patients [70]. Despite the evidence, although limited in some cases, of the clinical success of BRAF inhibitor monotherapy in BRAFV600E-mutant NSCLC, most patients developed adaptive resistance, resulting in reactivation of the MAPK pathway [71,72,73,74]. Several next-generation αC-helix IN RAF inhibitors, equipotent and dimer selective, including BGB-3245, BDTX-4933, exarafenib, and FORE8394, are currently under clinical evaluation as monotherapies for solid tumors bearing MAPK pathway mutations, including mutated NSCLC (Table 1, Figure 2) [75,76,77,78].

3.2. MEK Inhibitors as Monotherapy in NSCLC

Several clinical trials have investigated the role of MEK inhibitors in early clinical development for the treatment of advanced NSCLC. A phase II study compared the efficacy and safety of the MEK inhibitor AZD6244 versus the chemotherapy pemetrexed, and reported 5% and 4.5% ORRs for the MEK inhibitor-treated and chemotherapy-treated cohorts, respectively, but not significant difference in the median PFS [79]. In another phase II trial with the MEK inhibitor mirdametinib (PD-0325901), two intermittent administration schedules were tested but no objective responses were observed, and the median PFS was 1.8 months and the OS was 7.8 months [80]. Another phase II clinical trial comparing the MEK inhibitor, trametinib, to docetaxel, a microtubule depolymerization inhibitor in KRAS-mutant NSCLC patients, revealed no significant difference in survival outcomes [79]. However, early data from the “National Lung Matrix Trial”, testing among other targeted therapies selumetinib plus docetaxel, reported a confirmed OR for LUAD patients with NF1 loss [81]. A phase II basket trial tested selumetinib in molecularly profiled NSCLC patients harboring mostly KRAS and fewer BRAF, NRAS, and HRAS mutations. However, only one KRAS-mutant patient achieved a partial response with the other nine patients failing to meet the study’s primary endpoint [82]. In general, according to these clinical trials, monotherapy with MEK inhibitors seems to provide only modest survival benefits. However, there are newer compounds, ABM-168 and IMM-6-415, with increased potency for MEK, under preclinical and clinical testing, as monotherapies for MAPK-driven cancers, including lung cancer (Table 1) [83,84].

3.3. BRAF and MEK Inhibitor Combinations in NSCLC

3.3.1. BRAF-Mutant NSCLC

Following the clinical success from the combined use of BRAF and MEK inhibitors in BRAFV600E-mutant melanoma [52,85], the BRAF and MEK inhibitor combination was systematically evaluated in the context of BRAFV600E-mutant NSCLC. The favorable survival outcomes and safety profile of the combined dabrafenib plus trametinib treatment for BRAFV600E-mutant NSCLC led, in June 2017, to the FDA approval of the combination as both first and second lines, regardless of previous therapy. The approval was based on the results of a phase II clinical trial that evaluated the dabrafenib and trametinib combination in two cohorts: one treatment naïve and one with at least one prior therapy. Dabrafenib administration was evaluated as a single agent in a third cohort with at least one prior therapy [86,87]. All three cohorts exhibited favorable outcomes with the treatment-naive group showing the best responses, with an ORR of 64%, a 10.4-month DOR, and a median progression-free survival of 10.9 months [86]. Despite these positive outcomes, the combination of dabrafenib with trametinib is associated with an increased incidence of adverse effects [87]. Encorafenib is a more recently developed small molecule RAF monomer-selective inhibitor that exhibits an increased pharmacodynamic activity compared to vemurafenib or dabrafenib [88]. In 2018, the FDA granted approval for the combination of encorafenib with the MEK inhibitor binimetinib for the treatment of unresectable or metastatic melanoma patients bearing BRAF V600E/K mutations. This approval was based on the outcomes of the COLUMBUS trial (NCT01909453), a phase III randomized study involving 577 BRAF-mutant patients [53]. Patients who received the combination of encorafenib and binimetinib achieved a prolonged PFS compared to those treated with vemurafenib or encorafenib as monotherapy. Following that, positive results from the BEACON trial (NCT05456880) led to the FDA’s 2020 approval of the combination of encorafenib with the chimeric monoclonal anti-EGFR antibody cetuximab for the treatment of BRAFV600E-mutant patients with metastatic colorectal cancer [89]. Accordingly, the combination of encorafenib and binimetinib in both treatment-naive and previously treated BRAF-mutated NSCLC is currently being evaluated in several phase II trials (Table 1). In October 2023, the FDA granted approval to the combination of encorafenib with binimetinib for adult BRAFV600E-mutant NSCLC patients [90,91]. The efficacy of the combo was evaluated by the PHAROS clinical trial (NCT03915951, Table 1), an open-label, multicenter, single-arm study that enrolled 98 naïve-treated patients with metastatic BRAFV600E–mutated NSCLC. Preliminary findings from the PHAROS trial revealed an observed ORR of 75% in treatment-naive individuals and 46% in previously treated patients [90,91]. Another ongoing phase II clinical study (ENCO-BRAF trial; NCT04526782, Table 1) is evaluating the efficacy of the encorafenib–binimetinib combination in advanced BRAFV600E-mutant NSCLC patients. Furthermore, another phase II trial, testing the same combo, is enrolling metastatic NSCLC patients harboring BRAFV600E/K/D mutations (NCT03915951, Table 1). The primary endpoint of both studies is the ORR. The umbrella clinical trial, Landscape 1011 (NCT04585815, Table 2), is underway to assess the efficacy of sasanlimab, a novel anti-PD-1 monoclonal antibody administered subcutaneously, in combination with different targeted therapies in metastatic NSCLC patients. The first arm of the study includes BRAFV600E-mutant NSCLC patients who receive sasanlimab together with encorafenib plus binimetinib, with a durable ORR being the primary endpoint [92,93]. Key clinical trial outcomes for BRAF-mutant NSCLC patients upon treatment with RAF and/or MEK inhibitors, together with the most frequent adverse events, are presented in Table 3.

3.3.2. KRAS-Mutant NSCLC

Recently, the FDA has granted fast track designation to the RAF-MEK clamp inhibitor avutometinib plus the KRASG12C selective inhibitor sotorasib as a treatment for KRASG12C-mutant patients previously treated with one prior line of therapy excluding treatment with a KRASG12C inhibitor [95]. The combination of avutometinib plus sotorasib is being further evaluated in the phase I/II RAMP 203 study (NCT05074810, Table 1) [96]. Additionally, the combination of avutometinib with the mTOR inhibitor everolimus in KRAS-mutant NSCLC patients has shown promising results and is being tested in another clinical trial (NCT02407509, Table 1) [97].

3.3.3. EGFR-Mutant NSCLC

The MEK inhibitor selumetinib in combination with the third-generation EGFR tyrosine kinase inhibitor osmertinib has been evaluated, in parallel with the combinations of osimertinib plus savolitinib (MET inhibitor) or osimertinib plus durvalumab (anti-PD-L1 antibody), in a clinical trial investigating strategies to overcome EGFR inhibitor resistance (NCT02143466), in the context of EFGR-mutant NSCLC [98]. The ORRs were 42% for the selumetinib–osimertinib cohort, and 44% and 43% for the cohorts of osimertinib–savolitinib and osimertinib–durvalumab, respectively, but the study was discontinued due to increased reporting of interstitial lung disease in the osimertinib–durvalumab group [98]. An intermittent dosing of selumetinib in combination with osimertinib is currently under evaluation in a first-line phase II clinical study for advanced EFGR-mutant NSCLC patients (NCT03392246, Table 1).

4. BRAF and MEK Inhibitor-Mediated Resistance

Despite the clinical success of BRAF and MEK inhibitors, the development of adaptive and acquired drug resistance limits the effectiveness of the targeted therapy. Relief of negative feedback upon treatment with BRAF or MEK inhibitors is a major cause of adaptive drug resistance, as it results in RTK upregulation, RAS activation, and RAF dimerization promoting recovery of downstream ERK activity [10,57,99,100]. CRAF and ARAF paralogs, the other two RAF protein family members, apart from BRAF, have been identified as mediating resistance upon BRAF inhibitor treatment in melanoma [101]. Several mechanisms contributing to BRAF inhibitor-mediated acquired resistance may involve the formation of BRAF splice variants, BRAF gene amplification, and secondary activating mutations such as in RTKs, KRAS, NRAS, or MEK [102,103]. After vemurafenib treatment in BRAFV600E-mutant melanoma, the formation of p61BRAFV600E splice variants that can dimerize was found to drive the development of resistance [71]. The bypass activation of other associated signaling pathways can be another mechanism of adaptive resistance. Indeed, activation of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) pathway through RTK overexpression, phosphatase and tensin homolog (PTEN) loss of function, or activating mutations in phosphatidylinositol 3-kinase catalytic subunit (PI3KC) and AKT mediates the development of resistance to BRAF inhibition [104,105]. In BRAFV600-mutant NSCLC, resistance can occur after treatment with either BRAF inhibitors as single agents or upon dual BRAF and MEK inhibition by activating mutations in KRAS or NRAS [74,106,107]. These mutations include KRASG12D upon dabrafenib treatment and NRASQ61K upon dabrafenib and trametinib treatment. The clinical trial MATCH-R “Matching Resistance” identified that MEKK57N, KRASQ61R, and NRASQ61K mutations and a frameshift PTEN mutation in patients progressed to dual BRAF and MEK inhibition using dabrafenib and trametinib, respectively [108]. Another study reported, upon combined BRAF and MEK inhibition, transcriptional upregulation of fibroblast growth factor-1 (FGF-1), which resulted in autocrine fibroblast receptor (FGFR) activation, thus inducing MAPK pathway reactivation [109]. FGFR overexpression has been reported as a resistance mechanism to trametinib in KRAS-mutant NSCLC models [110]. Recently, elevated levels of cyclin-dependent kinase 4 (CDK4), a key cell cycle promoting protein, have been observed in a BRAFV600E-mutant NSCLC patient, following dabrafenib and trametinib co-treatment, and the ectopic expression of CDK4 in patient-derived BRAFV600E-mutant cells conferred partial resistance to dabrafenib [111]. Mutations in mixed lineage kinase 1 (MLK1) and Ras-related C3 botulinum toxin substrate 1 (RAC1) can lead to resistance mechanisms independent of RAF activation. Similarly, mutations in mitogen-activated protein kinase kinases 1 and 2 (MAP2K1 and MAP2K2), the genes that encode for MEK1 and MEK2, respectively, have been observed in both BRAFV600-mutant and BRAF non-V600-mutant tumors, with MEK-mutants exhibiting varying degrees of dependence on RAF for activation [112,113]. Certain MEK mutations, like MEK1 E102_I103del, confer resistance to allosteric MEK inhibitors but remain sensitive to ATP-competitive MEK inhibitors [114,115]. Downstream alterations, such as amplification of CCND1, which encodes for cyclin-D1 and loss of function of CDKN2A, which encodes for both p16INK4A and p14ARF, also contribute to resistance to RAF inhibitors [94,116,117,118]. Combinations of MEK or RAF inhibitors with CDK4/6 inhibitors have shown promise in preclinical and early clinical studies and are currently under further clinical investigation [Table 1 and Table 2] [94,118]. Thus, molecular profiling of co-occurring mutations is crucial for detecting possible acquired resistance mechanisms and guiding targeted therapy selection.

5. BRAF and MEK Inhibitor Toxicities

The therapeutic benefits of RAF and MEK inhibitors in the management of NSCLC come with a spectrum of toxicities and adverse events that necessitate careful consideration. Common toxicities associated with RAF inhibitors include cutaneous reactions such as rash and photosensitivity, often requiring dose adjustments or temporary treatment interruptions, while hypertension, pyrexia, and increased liver enzymes are the most common adverse events [119,120,121]. Additionally, MEK inhibitors are known to induce gastrointestinal toxicities, including diarrhea, nausea, and vomiting, or may cause interstitial lung disease (Table 3) [120]. Although these side effects are generally manageable, they can impact patients’ quality of life and treatment compliance. Collaborative efforts are essential to comprehensively understand the toxicological profiles and devise strategies for mitigating adverse events to optimize the therapeutic potential of RAF and MEK inhibitors in NSCLC treatment. Ongoing research and clinical trials aim to refine the use of these agents, offering a balance between efficacy and tolerability to improve patient outcomes.

6. Discussion

The successful implementation of immune checkpoint inhibitors (ICIs) in the management of melanoma and other cancers has directed efforts to targeted therapy/immunotherapy approaches. Thus far, these attempts have been largely fruitless due to overlapping toxicities that led to early treatment termination [122,123]. Despite that, a triple combination of atezolizumab, an anti-PD-L1 monoclonal antibody, with vemurafenib plus cobimetinib was granted FDA approval in 2020 as a first-line therapy option for patients with advanced BRAFV600E-mutant melanoma [124]. Several ongoing clinical trials are evaluating the combination of BRAF and/or MEK inhibitors together with immune checkpoint inhibitors (ICIs) (Table 1 and Table 2). This strategy is supported by preclinical evidence indicating a synergistic activity of MEK inhibition with immune checkpoint blockade. The biological rationale of this approach relies on the observed immunomodulatory effects of MAPK inhibition in the tumor microenvironment, such as the transcriptional decrease in PD-L1 expression, upregulation of MHC-I expression in tumor cells, and increased pro-inflammatory cytokine production [125,126]. In addition, the RAF-MEK clamp inhibitor avutometinib has been shown, in preclinical models, to induce the antitumor activity of ICIs [127]. Thus, a clinical trial is testing the efficacy and tolerability of the triple combination of the MEK inhibitor selumetinib, in continuous or intermittent dosing schedules, plus durvalumab, an anti-PD-L1, and tremelimumab, an anti-CTLA4, as antibodies in advanced or metastatic NSCLC patients (NCT03581487, Table 2) [128]. Furthermore, this approach is supported by the observed PD-L1 expression in BRAF-mutants and by the enhanced T cell-mediated immunity from BRAF and MEK inhibitors. There is evidence suggesting that BRAF and MEK inhibition induces CD4 and CD8 T lymphocytes, boosting their cytotoxicity against cancer cells, which is evident by elevated granzyme B and perforin levels, and an elevated expression of cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) [111,129,130,131,132,133]. Targeting of CTLA-4 together with MEK, using trametinib or selumetinib, resulted in increased survival in KRAS-mutant lung cancer mouse models [134,135]. In an advanced BRAFV600E-mutant NSCLC case study, the combination of atezolizumab and platinum-based chemotherapy led to a prolonged response [135]. The combination of atezolizumab and cobimetinib has been further investigated in a trial with various solid tumors, in which the NSCLC cohort had an ORR of 18% and a median OS of 13.2 months, with a 12-month OS of 57% [136]. An ongoing multicohort clinical study is testing, among other treatments, the triple combination of atezolizumab plus vemurafenib and cobimetinib in BRAFV600E-mutant NSCLC patients. The early data of the triple combo suggest further investigation (NCT03178552, Table 2).
While new combinatorial strategies may help in addressing the development of therapeutic resistance, the advancement in the medicinal chemistry field provides many new small molecule inhibitors and targeting agents, like Proteolysis Targeted Chimeras (PROTACs), with unique and improved biochemical properties that may provide better outcomes. For instance, the use of the next-generation type 1.5 (αC-helix OUT) RAF inhibitor PLX8394, which abolishes the paradoxical ERK activation “paradox-breaker”, prevented the BRAF inhibitor monotherapy-mediated drug resistance in BRAFV600E-mutant NSCLC [137]. Moreover, the same compound had additional preclinical activity in certain BRAF non-V600 NSCLC models [137], indicating that a RAF inhibitor with distinct biochemical characteristics can provide additional benefits. HLX208 is another, more recent, novel RAF monomer-selective inhibitor that has shown promising efficacy and a well-tolerated safety profile in BRAFV600E-mutant adult patients with Langerhans cell histiocytosis (LCH) and/or Erdheim–Chester disease (ECD) [138,139]. Currently, HLX208 is being evaluated in a clinical trial of solid tumors in combination with trametinib (NCT04965220, Table 1).
In a different strategy, the concept of implementing a prolonged on–off schedule or introducing “drug holidays” has been suggested as a strategy to favor the expansion of sensitive clones to targeted therapy over resistant ones, thereby maintaining tumor responsiveness [140,141,142]. A similar approach has been proposed in a recent case of successful re-challenge therapy with combined BRAF and MEK inhibition following a series of subsequent chemotherapy treatments. The response mechanism may involve the establishment of a “drug-free” environment, leading to a decrease in the number of heterogeneous tumor cells previously exposed to BRAF and MEK inhibitors, ultimately resulting in a successful re-challenge for certain patients [143]. Downstream inhibition of the terminal pathway kinase, ERK, represents a promising approach to suppress any upstream pathway activation or reactivation. Currently, there are several ERK inhibitors under preclinical and clinical evaluation for MAPK-driven cancers [144,145].
Autophagy, a catabolic cellular process that replenishes nutrients and restores damaged organelles, has been identified as s resistance mechanism to combined BRAF and MEK inhibition in melanoma [146]. Targeting autophagy together with BRAF and MEK inhibitors has provided clinical benefit for advanced BRAFV600E-mutant melanoma [147,148]. Unc-51-like kinase (ULK) kinase integrates inputs from nutrient and stress sensors to initiate autophagy [146]. DCC-3116 is a potent and selective inhibitor of ULK that has exhibited preclinical evidence and is now under clinical evaluation in combination with trametinib, binimetinib, or sotorasib, a KRASG12C inhibitor, for MAPK-driven cancer (NC04892017, Table 2) [149]. Alternatively, co-targeting pyroptosis, an alternative type of non-apoptotic, inflammation-related programmed cell death, with MAPK pathway inhibitors seems an attractive approach [150,151]. This is supported by the absence of pyroptosis-related markers in BRAF and MEK-combined inhibition-resistant BRAF-mutant cancer models, which was associated with reduced T cell antitumor immunity and, importantly, was sensitive to pyroptosis-inducing chemotherapy [152].
SHP2 phosphatase has been an attractive antitumor target, mainly because of its key role as a signal transduction node downstream of multiple RTKs and, subsequently, as a positive regulator of RAS and MAPK signaling [153,154,155]. More specifically, SHP2 inhibitors have shown efficacy against tumors harboring class III BRAF mutations, certain KRAS mutations, such as KRASG12C, EGFR mutations, or with NF1 loss [156,157,158]. However, resistance to single-agent SHP2 inhibition or combination with MEK inhibition has been observed in cases of KRASG13D and KRASQ61 mutations, and some BRAFV600E models. This resistance was attributed to FGFR-mediated, SHP2-independent feedback reactivation of RAS signaling pathways [156]. In a first-in-human trial of various tumor models, including BRAFV600E-mutant colorectal cancer and KRASG12D-mutant ovarian cancer, after disease progression on monotherapy with PF-07284892, an allosteric SHP2 inhibitor, a combination of encorafenib with binimetinib was administered in addition to PF-07284892. The triple combination therapy led to tumor response and to prolongation of the overall clinical benefit [159,160]. This clinical study provides a rationale for the utility of RAF and MEK inhibitors in combination with SHP2 inhibitors in overcoming resistance to targeted therapies, presenting a valuable model for testing novel drug combinations early in clinical development.

6.1. Future Directions—Next-Generation RAF Inhibitors

There is preclinical evidence that next-generation αC-helix IN RAF inhibitors, and more specifically, the subgroup of RAF dimer selective inhibitors, may provide benefits in targeting BRAF non-V600 mutants (class II and III BRAF mutations) in NSCLC [43]. However, in the absence of comprehensive studies, the current guidelines for clinicians suggest that BRAF non-V600E mutants should be managed as non-mutationally-driven NSCLC [161]. Naporafenib, a RAF dimer selective inhibitor, in combination with the MEK inhibitor trametinib or in combination with the ERK inhibitor LTT462, is being explored in a clinical trial for its efficacy in advanced metastatic KRAS- or BRAF-mutant NSCLC or in NRAS-mutant melanoma (NCT02974725, Table 1). Preliminary results from the NRAS-mutant cohort demonstrated a promising ORR of 47% for the first 15 enrolled patients [162]. Lifirafenib (BGB-283), another RAF inhibitor of the same category (αC-IN, equipotent for RAF monomers and dimer), has shown antitumor activity in KRAS-mutant NSCLC [70]. Currently, lifirafenib in combination with the MEK inhibitor mirdametinib is being evaluated in a phase I clinical trial (NCT03905148, Table 1) with preliminary findings indicating a favorable safety profile and showing evidence of antitumor activity, including in NSCLC patients harboring KRAS, NRAS, and BRAF mutations. [147]. These results set the base for further exploration of these next-generation RAF inhibitors beyond BRAF-mutant NSCLC. In a phase Ib trial, belvarafenib, another dimer selective RAF inhibitor, in combination with the MEK inhibitor cobimetinib, demonstrated a partial response in two BRAF non-V600-mutant NSCLC patients tested [163]. An ongoing phase I multicenter study is testing belvarafenib, in combination with cobimetinib or with the anti-EGFR monoclonal antibody cetuximab, in locally advanced or metastatic solid tumors (NCT03284502, Table 1).

6.2. Biomarkers—Predictors of Response to RAF and MEK Inhibitors

The National Comprehensive Cancer Network (NCCN) NSCLC panel currently recommends BRAF mutation testing for of all patients with metastatic non-squamous NSCLC and suggests the combination targeted therapy of dabrafenib plus trametinib as a preferred first-line therapy for BRAFV600E-mutant NSCLC patients [164]. Drawing insights from recent studies, predictive biomarkers offer valuable insights into treatment response and outcomes. The first study highlights the potential of specific biomarkers, including p27 accumulation, cyclin D1 downregulation, PARP cleavage, and increased phospho-4E-BP1, in predicting cellular responses to trametinib treatment, indicating a spectrum of outcomes ranging from apoptosis to autophagy, providing a framework for understanding treatment efficacy [165]. Moreover, insights from a more recent study underscore the limitations of combined RAF and MEK inhibition in NSCLC patients with activated BRAF non-V600 mutations. Despite well-tolerated treatment regimens, incomplete ERK pathway inhibition was observed in on-treatment biopsies, suggesting a need for more robust predictive biomarkers to identify patients likely to benefit from this therapeutic approach [166]. Moreover, the recent association of phenotypic and spatial characteristics of cancer-associated fibroblasts (CAFs) in the tumor microenvironment of NSCLC, with patient outcome [167], is worth exploring further in the context of RAF and MEK inhibitor targeted therapies in BRAF-mutant NSCLC. Overall, these findings underscore the importance of a comprehensive molecular profiling and personalized medicine in NSCLC management, emphasizing the critical role of predictive biomarkers in optimizing treatment strategies and improving patient outcomes.

7. Conclusions—Future Perspectives

The ongoing advancements in the development of the next generation of RAF and MEK inhibitors, characterized by distinct biochemical properties, present promising avenues for the treatment of lung cancer. These novel inhibitors are currently undergoing rigorous preclinical and clinical evaluations. Their exploration offers a potential solution to the challenges posed by adaptive resistance and the applicability of existing inhibitors to specific mutational profiles, such as BRAFV600E-mutant NSCLC patients. Moreover, the evolving landscape of precision medicine and targeted therapies continues to fuel optimism for overcoming these challenges. In conclusion, while the approvals of the dabrafenib–trametinib and encorafenib–binimetinib combinations represent significant strides in the treatment of BRAFV600E-mutated NSCLC, the field is actively working towards more inclusive and effective therapeutic strategies. Improved targeting agents moving into the clinic and rational combinatorial strategies, targeting selectively key components “inside” and “outside” the MAPK pathway, have become more beneficial. The very recently granted fast track designation for the combination of the RAF−MEK clamp inhibitor avutometinib plus the KRASG12C selective inhibitor sotorasib for the treatment of KRASG12C-mutant NSCLC patients underscores the continuing potential of RAF and MEK inhibition against RAS/MAPK-driven malignancies. The ongoing research on next-generation inhibitors and the expansion of targetable mutations hold promise for a more comprehensive and adaptable approach to address drug resistance and widen the therapeutic impact of RAF and MEK inhibition in the diverse landscape of lung cancer.

Author Contributions

Conceptualization, C.A., P.I.P. and A.G.P.; writing—original draft preparation, C.A. and K.A.P.; literature search, C.A. and K.A.P.; supervision, A.G.P.; writing—review and editing, P.I.P. and A.G.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Kratzer, T.B.; Bandi, P.; Freedman, N.D.; Smith, R.A.; Travis, W.D.; Jemal, A.; Siegel, R.L. Lung cancer statistics, 2023. Cancer 2023, 130, 1330–1348. [Google Scholar] [CrossRef]
  2. LoPiccolo, J.; Gusev, A.; Christiani, D.C.; Jänne, P.A. Lung cancer in patients who have never smoked—An emerging disease. Nat. Rev. Clin. Oncol. 2024, 21, 121–146. [Google Scholar] [CrossRef]
  3. Siegel, R.L.; Giaquinto, A.N.; Jemal, A. Cancer statistics, 2024. CA A Cancer J. Clin. 2024, 74, 12–49. [Google Scholar] [CrossRef]
  4. Travis, W.D.; Brambilla, E.; Nicholson, A.G.; Yatabe, Y.; Austin, J.H.M.; Beasley, M.B.; Chirieac, L.R.; Dacic, S.; Duhig, E.; Flieder, D.B.; et al. The 2015 World Health Organization Classification of Lung Tumors: Impact of Genetic, Clinical and Radiologic Advances Since the 2004 Classification. J. Thorac. Oncol. 2015, 10, 1243–1260. [Google Scholar] [CrossRef]
  5. Schabath, M.B.; Cote, M.L. Cancer Progress and Priorities: Lung Cancer. Cancer Epidemiol. Biomark. Prev. 2019, 28, 1563–1579. [Google Scholar] [CrossRef]
  6. Herbst, R.S.; Morgensztern, D.; Boshoff, C. The biology and management of non-small cell lung cancer. Nature 2018, 553, 446–454. [Google Scholar] [CrossRef]
  7. Wiesweg, M.; Kasper, S.; Worm, K.; Herold, T.; Reis, H.; Sara, L.; Metzenmacher, M.; Abendroth, A.; Darwiche, K.; Aigner, C.; et al. Impact of RAS mutation subtype on clinical outcome-a cross-entity comparison of patients with advanced non-small cell lung cancer and colorectal cancer. Oncogene 2019, 38, 2953–2966. [Google Scholar] [CrossRef]
  8. Mayer, C.; Ofek, E.; Fridrich, D.E.; Molchanov, Y.; Yacobi, R.; Gazy, I.; Hayun, I.; Zalach, J.; Paz-Yaacov, N.; Barshack, I. Direct identification of ALK and ROS1 fusions in non-small cell lung cancer from hematoxylin and eosin-stained slides using deep learning algorithms. Mod. Pathol. 2022, 35, 1882–1887. [Google Scholar] [CrossRef]
  9. Relli, V.; Trerotola, M.; Guerra, E.; Alberti, S. Abandoning the Notion of Non-Small Cell Lung Cancer. Trends Mol. Med. 2019, 25, 585–594. [Google Scholar] [CrossRef]
  10. Poulikakos, P.I.; Sullivan, R.J.; Yaeger, R. Molecular Pathways and Mechanisms of BRAF in Cancer Therapy. Clin. Cancer Res. 2022, 28, 4618–4628. [Google Scholar] [CrossRef]
  11. Hanrahan, A.J.; Chen, Z.; Rosen, N.; Solit, D.B. BRAF—A Tumour-Agnostic Drug Target with Lineage-Specific Dependencies. Nat. Rev. Clin. Oncol. 2024, 21, 224–247. [Google Scholar] [CrossRef]
  12. Lavoie, H.; Therrien, M. Regulation of RAF protein kinases in ERK signalling. Nat. Rev. Mol. Cell Biol. 2015, 16, 281–298. [Google Scholar] [CrossRef]
  13. Karoulia, Z.; Gavathiotis, E.; Poulikakos, P.I. New perspectives for targeting RAF kinase in human cancer. Nat. Rev. Cancer 2017, 17, 676–691. [Google Scholar] [CrossRef]
  14. Tran, T.H.; Chan, A.H.; Young, L.C.; Bindu, L.; Neale, C.; Messing, S.; Dharmaiah, S.; Taylor, T.; Denson, J.P.; Esposito, D.; et al. KRAS interaction with RAF1 RAS-binding domain and cysteine-rich domain provides insights into RAS-mediated RAF activation. Nat. Commun. 2021, 12, 1176. [Google Scholar] [CrossRef]
  15. Karoulia, Z.; Wu, Y.; Ahmed, T.A.; Xin, Q.; Bollard, J.; Krepler, C.; Wu, X.; Zhang, C.; Bollag, G.; Herlyn, M.; et al. An Integrated Model of RAF Inhibitor Action Predicts Inhibitor Activity against Oncogenic BRAF Signaling. Cancer Cell 2016, 30, 485–498. [Google Scholar] [CrossRef]
  16. Moore, A.R.; Rosenberg, S.C.; McCormick, F.; Malek, S. RAS-targeted therapies: Is the undruggable drugged? Nat. Rev. Drug Discov. 2020, 19, 533–552. [Google Scholar] [CrossRef]
  17. Caunt, C.J.; Sale, M.J.; Smith, P.D.; Cook, S.J. MEK1 and MEK2 inhibitors and cancer therapy: The long and winding road. Nat. Rev. Cancer 2015, 15, 577–592. [Google Scholar] [CrossRef]
  18. Luk, P.P.; Yu, B.; Ng, C.C.; Mercorella, B.; Selinger, C.; Lum, T.; Kao, S.; O’toole, S.A.; Cooper, W.A. BRAF mutations in non-small cell lung cancer. Transl. Lung Cancer Res. 2015, 4, 142–148. [Google Scholar] [CrossRef]
  19. Dang, C.V. MYC on the Path to Cancer. Cell 2012, 149, 22–35. [Google Scholar] [CrossRef]
  20. Pratilas, C.A.; Taylor, B.S.; Ye, Q.; Viale, A.; Sander, C.; Solit, D.B.; Rosen, N. V600E BRAF is associated with disabled feedback inhibition of RAF–MEK signaling and elevated transcriptional output of the pathway. Proc. Natl. Acad. Sci. USA 2009, 106, 4519–4524. [Google Scholar] [CrossRef]
  21. Dearden, S.; Stevens, J.; Wu, Y.L.; Blowers, D. Mutation incidence and coincidence in non small-cell lung cancer: Meta-analyses by ethnicity and histology (mutMap). Ann. Oncol. 2013, 24, 2371–2376. [Google Scholar] [CrossRef]
  22. Grosse, A.; Grosse, C.; Rechsteiner, M.; Soltermann, A. Analysis of the frequency of oncogenic driver mutations and correlation with clinicopathological characteristics in patients with lung adenocarcinoma from Northeastern Switzerland. Diagn. Pathol. 2019, 14, 18. [Google Scholar] [CrossRef]
  23. Mendiratta, G.; Ke, E.; Aziz, M.; Liarakos, D.; Tong, M.; Stites, E.C. Cancer gene mutation frequencies for the U.S. population. Nat. Commun. 2021, 12, 5961. [Google Scholar] [CrossRef]
  24. Davies, H.; Bignell, G.R.; Cox, C.; Stephens, P.; Edkins, S.; Clegg, S.; Teague, J.; Woffendin, H.; Garnett, M.J.; Bottomley, W.; et al. Mutations of the BRAF gene in human cancer. Nature 2002, 417, 949–954. [Google Scholar] [CrossRef]
  25. A Hall-Jackson, C.; A Eyers, P.; Cohen, P.; Goedert, M.; Boyle, F.T.; Hewitt, N.; Plant, H.; Hedge, P. Paradoxical activation of Raf by a novel Raf inhibitor. Chem. Biol. 1999, 6, 559–568. [Google Scholar] [CrossRef]
  26. Mangana, J.; Levesque, M.P.; Karpova, M.B.; Dummer, R. Sorafenib in melanoma. Expert Opin. Investig. Drugs 2012, 21, 557–568. [Google Scholar] [CrossRef]
  27. Roskoski, R., Jr. Classification of small molecule protein kinase inhibitors based upon the structures of their drug-enzyme complexes. Pharmacol. Res. 2016, 103, 26–48. [Google Scholar] [CrossRef]
  28. Bollag, G.; Hirth, P.; Tsai, J.; Zhang, J.; Ibrahim, P.N.; Cho, H.; Spevak, W.; Zhang, C.; Zhang, Y.; Habets, G.; et al. Clinical efficacy of a RAF inhibitor needs broad target blockade in BRAF-mutant melanoma. Nature 2010, 467, 596–599. [Google Scholar] [CrossRef]
  29. Chapman, P.B.; Hauschild, A.; Robert, C.; Haanen, J.B.; Ascierto, P.; Larkin, J.; Dummer, R.; Garbe, C.; Testori, A.; Maio, M.; et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N. Engl. J. Med. 2011, 364, 2507–2516. [Google Scholar] [CrossRef]
  30. Tsai, J.; Lee, J.T.; Wang, W.; Zhang, J.; Cho, H.; Mamo, S.; Bremer, R.; Gillette, S.; Kong, J.; Haass, N.K.; et al. Discovery of a selective inhibitor of oncogenic B-Raf kinase with potent antimelanoma activity. Proc. Natl. Acad. Sci. USA 2008, 105, 3041–3046. [Google Scholar] [CrossRef]
  31. Heidorn, S.J.; Milagre, C.; Whittaker, S.; Nourry, A.; Niculescu-Duvas, I.; Dhomen, N.; Hussain, J.; Reis-Filho, J.S.; Springer, C.J.; Pritchard, C.; et al. Kinase-Dead BRAF and Oncogenic RAS Cooperate to Drive Tumor Progression through CRAF. Cell 2010, 140, 209–221. [Google Scholar] [CrossRef]
  32. Poulikakos, P.I.; Zhang, C.; Bollag, G.; Shokat, K.M.; Rosen, N. RAF inhibitors transactivate RAF dimers and ERK signalling in cells with wild-type BRAF. Nature 2010, 464, 427–430. [Google Scholar] [CrossRef]
  33. Hatzivassiliou, G.; Song, K.; Yen, I.; Brandhuber, B.J.; Anderson, D.J.; Alvarado, R.; Ludlam, M.J.C.; Stokoe, D.; Gloor, S.L.; Vigers, G.; et al. RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth. Nature 2010, 464, 431–435. [Google Scholar] [CrossRef]
  34. Yao, Z.; Yaeger, R.; Rodrik-Outmezguine, V.S.; Tao, A.; Torres, N.M.; Chang, M.T.; Drosten, M.; Zhao, H.; Cecchi, F.; Hembrough, T.; et al. Tumours with class 3 BRAF mutants are sensitive to the inhibition of activated RAS. Nature 2017, 548, 234–238. [Google Scholar] [CrossRef]
  35. Yao, Z.; Torres, N.M.; Tao, A.; Gao, Y.; Luo, L.; Li, Q.; de Stanchina, E.; Abdel-Wahab, O.; Solit, D.B.; Poulikakos, P.I.; et al. BRAF Mutants Evade ERK-Dependent Feedback by Different Mechanisms that Determine Their Sensitivity to Pharmacologic Inhibition. Cancer Cell 2015, 28, 370–383. [Google Scholar] [CrossRef]
  36. Tang, Z.; Yuan, X.; Du, R.; Cheung, S.H.; Zhang, G.; Wei, J.; Zhao, Y.; Feng, Y.; Peng, H.; Zhang, Y.; et al. BGB-283, a Novel RAF Kinase and EGFR Inhibitor, Displays Potent Antitumor Activity in BRAF-Mutated Colorectal Cancers. Mol. Cancer Ther. 2015, 14, 2187–2197. [Google Scholar] [CrossRef]
  37. Nakamura, A.; Arita, T.; Tsuchiya, S.; Donelan, J.; Chouitar, J.; Carideo, E.; Galvin, K.; Okaniwa, M.; Ishikawa, T.; Yoshida, S. Antitumor Activity of the Selective Pan-RAF Inhibitor TAK-632 in BRAF Inhibitor-Resistant Melanoma. Cancer Res 2013, 73, 7043–7055. [Google Scholar] [CrossRef]
  38. Chen, S.H.; Zhang, Y.; Van Horn, R.D.; Yin, T.; Buchanan, S.; Yadav, V.; Mochalkin, I.; Wong, S.S.; Yue, Y.G.; Huber, L.; et al. LY3009120. Cancer Discov. 2016, 6, 300–315. [Google Scholar] [CrossRef]
  39. Peng, S.B.; Henry, J.R.; Kaufman, M.D.; Lu, W.-P.; Smith, B.D.; Vogeti, S.; Rutkoski, T.J.; Wise, S.; Chun, L.; Zhang, Y.; et al. Inhibition of RAF Isoforms and Active Dimers by LY3009120 Leads to Anti-tumor Activities in RAS or BRAF Mutant Cancers. Cancer Cell 2015, 28, 384–398. [Google Scholar] [CrossRef]
  40. Adamopoulos, C.; Ahmed, T.A.; Tucker, M.R.; Ung, P.M.; Xiao, M.; Karoulia, Z.; Amabile, A.; Wu, X.; Aaronson, S.A.; Ang, C.; et al. Exploiting Allosteric Properties of RAF and MEK Inhibitors to Target Therapy-Resistant Tumors Driven by Oncogenic BRAF Signaling. Cancer Discov. 2021, 11, 1716–1735. [Google Scholar] [CrossRef]
  41. Puri, M.; Gawri, K.; Dawar, R. Therapeutic strategies for BRAF mutation in non-small cell lung cancer: A review. Front. Oncol. 2023, 13, 1141876. [Google Scholar] [CrossRef]
  42. Sforza, V.; Palumbo, G.; Cascetta, P.; Carillio, G.; Manzo, A.; Montanino, A.; Sandomenico, C.; Costanzo, R.; Esposito, G.; Laudato, F.; et al. BRAF Inhibitors in Non-Small Cell Lung Cancer. Cancers 2022, 14, 4863. [Google Scholar] [CrossRef]
  43. Negrao, M.V.; Raymond, V.M.; Lanman, R.B.; Robichaux, J.P.; He, J.; Nilsson, M.B.; Ng, P.K.; Amador, B.E.; Roarty, E.B.; Nagy, R.J.; et al. Molecular Landscape of BRAF-Mutant NSCLC Reveals an Association Between Clonality and Driver Mutations and Identifies Targetable Non-V600 Driver Mutations. J. Thorac. Oncol. 2020, 15, 1611–1623. [Google Scholar] [CrossRef]
  44. Dagogo-Jack, I.; Martinez, P.; Yeap, B.Y.; Ambrogio, C.; Ferris, L.A.; Lydon, C.; Nguyen, T.; Jessop, N.A.; Iafrate, A.J.; Johnson, B.E.; et al. Impact of BRAF Mutation Class on Disease Characteristics and Clinical Outcomes in BRAF-mutant Lung Cancer. Clin. Cancer Res. 2019, 25, 158–165. [Google Scholar] [CrossRef]
  45. Dudnik, E.; Peled, N.; Nechushtan, H.; Wollner, M.; Onn, A.; Agbarya, A.; Moskovitz, M.; Keren, S.; Popovits-Hadari, N.; Urban, D.; et al. BRAF Mutant Lung Cancer: Programmed Death Ligand 1 Expression, Tumor Mutational Burden, Microsatellite Instability Status, and Response to Immune Check-Point Inhibitors. J. Thorac. Oncol. 2018, 13, 1128–1137. [Google Scholar] [CrossRef]
  46. Venkatanarayan, A.; Liang, J.; Yen, I.; Shanahan, F.; Haley, B.; Phu, L.; Verschueren, E.; Hinkle, T.B.; Kan, D.; Segal, E.; et al. CRAF dimerization with ARAF regulates KRAS-driven tumor growth. Cell Rep. 2022, 38, 110351. [Google Scholar] [CrossRef]
  47. Planchard, D.; Kim, T.M.; Mazieres, J.; Quoix, E.; Riely, G.; Barlesi, F.; Souquet, P.-J.; Smit, E.F.; Groen, H.J.M.; Kelly, R.J.; et al. Dabrafenib in patients with BRAFV600E-positive advanced non-small-cell lung cancer: A single-arm, multicentre, open-label, phase 2 trial. Lancet Oncol. 2016, 17, 642–650. [Google Scholar] [CrossRef]
  48. Zhao, Y.; Adjei, A.A. The clinical development of MEK inhibitors. Nat. Rev. Clin. Oncol. 2014, 11, 385–400. [Google Scholar] [CrossRef]
  49. Robert, C.; Flaherty, K.; Nathan, P.; Hersey, P.; Garbe, C.; Milhem, M.; Demidov, L.; Mohr, P.; Hassel, J.C.; Rutkowski, P.; et al. Five-year outcomes from a phase 3 METRIC study in patients with BRAF V600 E/K–mutant advanced or metastatic melanoma. Eur. J. Cancer 2019, 109, 61–69. [Google Scholar] [CrossRef]
  50. Long, G.V.; Hauschild, A.; Santinami, M.; Atkinson, V.; Mandalà, M.; Sileni, V.C.; Larkin, J.; Nyakas, M.; Dutriaux, C.; Haydon, A.; et al. Adjuvant Dabrafenib plus Trametinib in Stage IIIBRAF-Mutated Melanoma. N. Engl. J. Med. 2017, 377, 1813–1823. [Google Scholar] [CrossRef]
  51. Gross, A.M.; Wolters, P.L.; Dombi, E.; Baldwin, A.; Whitcomb, P.; Fisher, M.J.; Weiss, B.; Kim, A.; Bornhorst, M.; Shah, A.C.; et al. Selumetinib in Children with Inoperable Plexiform Neurofibromas. N. Engl. J. Med. 2020, 382, 1430–1442. [Google Scholar] [CrossRef]
  52. Ascierto, P.A.; McArthur, G.A.; Dréno, B.; Atkinson, V.; Liszkay, G.; Di Giacomo, A.M.; Mandalà, M.; Demidov, L.; Stroyakovskiy, D.; Thomas, L.; et al. Cobimetinib combined with vemurafenib in advanced BRAFV600-mutant melanoma (coBRIM): Updated efficacy results from a randomised, double-blind, phase 3 trial. Lancet Oncol. 2016, 17, 1248–1260. [Google Scholar] [CrossRef]
  53. Dummer, R.; Ascierto, P.A.; Gogas, H.J.; Arance, A.; Mandala, M.; Liszkay, G.; Garbe, C.; Schadendorf, D.; Krajsova, I.; Gutzmer, R.; et al. Encorafenib plus binimetinib versus vemurafenib or encorafenib in patients with BRAF -mutant melanoma (COLUMBUS): A multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 2018, 19, 603–615. [Google Scholar] [CrossRef]
  54. Ishii, N.; Harada, N.; Joseph, E.W.; Ohara, K.; Miura, T.; Sakamoto, H.; Matsuda, Y.; Tomii, Y.; Tachibana-Kondo, Y.; Iikura, H.; et al. Enhanced Inhibition of ERK Signaling by a Novel Allosteric MEK Inhibitor, CH5126766, That Suppresses Feedback Reactivation of RAF Activity. Cancer Res. 2013, 73, 4050–4060. [Google Scholar] [CrossRef]
  55. Lito, P.; Saborowski, A.; Yue, J.; Solomon, M.; Joseph, E.; Gadal, S.; Saborowski, M.; Kastenhuber, E.; Fellmann, C.; Ohara, K.; et al. Disruption of CRAF-Mediated MEK Activation Is Required for Effective MEK Inhibition in KRAS Mutant Tumors. Cancer Cell 2014, 25, 697–710. [Google Scholar] [CrossRef]
  56. Hatzivassiliou, G.; Haling, J.R.; Chen, H.; Song, K.; Price, S.; Heald, R.; Hewitt, J.F.M.; Zak, M.; Peck, A.; Orr, C.; et al. Mechanism of MEK inhibition determines efficacy in mutant KRAS- versus BRAF-driven cancers. Nature 2013, 501, 232–236. [Google Scholar] [CrossRef]
  57. Yaeger, R.; Corcoran, R.B. Targeting Alterations in the RAF–MEK Pathway. Cancer Discov. 2019, 9, 329–341. [Google Scholar] [CrossRef]
  58. Marks, J.L.; Gong, Y.; Chitale, D.; Golas, B.; McLellan, M.D.; Kasai, Y.; Ding, L.; Mardis, E.R.; Wilson, R.K.; Solit, D.; et al. Novel MEK1 Mutation Identified by Mutational Analysis of Epidermal Growth Factor Receptor Signaling Pathway Genes in Lung Adenocarcinoma. Cancer Res. 2008, 68, 5524–5528. [Google Scholar] [CrossRef]
  59. Arcila, M.E.; Drilon, A.; Sylvester, B.E.; Lovly, C.M.; Borsu, L.; Reva, B.; Kris, M.G.; Solit, D.B.; Ladanyi, M. MAP2K1 (MEK1) Mutations Define a Distinct Subset of Lung Adenocarcinoma Associated with Smoking. Clin. Cancer Res. 2015, 21, 1935–1943. [Google Scholar] [CrossRef]
  60. Sosman, J.A.; Kim, K.B.; Schuchter, L.; Gonzalez, R.; Pavlick, A.C.; Weber, J.S.; McArthur, G.A.; Hutson, T.E.; Moschos, S.J.; Flaherty, K.T.; et al. Survival in BRAF V600–Mutant Advanced Melanoma Treated with Vemurafenib. N. Engl. J. Med. 2012, 366, 707–714. [Google Scholar] [CrossRef]
  61. Flaherty, K.T.; Puzanov, I.; Kim, K.B.; Ribas, A.; McArthur, G.A.; Sosman, J.A.; O’Dwyer, P.J.; Lee, R.J.; Grippo, J.F.; Nolop, K.; et al. Inhibition of Mutated, Activated BRAF in Metastatic Melanoma. N. Engl. J. Med. 2010, 363, 809–819. [Google Scholar] [CrossRef]
  62. Peters, S.; Michielin, O.; Zimmermann, S. Dramatic Response Induced by Vemurafenib in a BRAF V600E-Mutated Lung Adenocarcinoma. J. Clin. Oncol. 2013, 31, e341–e344. [Google Scholar] [CrossRef]
  63. Gautschi, O.; Pauli, C.; Strobel, K.; Hirschmann, A.; Printzen, G.; Aebi, S.; Diebold, J. A Patient with BRAF V600E Lung Adenocarcinoma Responding to Vemurafenib. J. Thorac. Oncol. 2012, 7, e23–e24. [Google Scholar] [CrossRef]
  64. Gautschi, O.; Milia, J.; Cabarrou, B.; Bluthgen, M.V.; Besse, B.; Smit, E.F.; Wolf, J.; Peters, S.; Früh, M.; Koeberle, D.; et al. Targeted Therapy for Patients with BRAF-Mutant Lung Cancer: Results from the European EURAF Cohort. J. Thorac. Oncol. 2015, 10, 1451–1457. [Google Scholar] [CrossRef]
  65. Hyman, D.M.; Puzanov, I.; Subbiah, V.; Faris, J.E.; Chau, I.; Blay, J.-Y.; Wolf, J.; Raje, N.S.; Diamond, E.L.; Hollebecque, A.; et al. Vemurafenib in Multiple Nonmelanoma Cancers with BRAF V600 Mutations. N. Engl. J. Med. 2015, 373, 726–736. [Google Scholar] [CrossRef]
  66. Mazieres, J.; Cropet, C.; Montané, L.; Barlesi, F.; Souquet, P.; Quantin, X.; Dubos-Arvis, C.; Otto, J.; Favier, L.; Avrillon, V.; et al. Vemurafenib in non-small-cell lung cancer patients with BRAFV600 and BRAFnonV600 mutations. Ann. Oncol. 2020, 31, 289–294. [Google Scholar] [CrossRef]
  67. Robinson, S.D.; O’Shaughnessy, J.A.; Cowey, C.L.; Konduri, K. BRAF V600E-mutated lung adenocarcinoma with metastases to the brain responding to treatment with vemurafenib. Lung Cancer 2014, 85, 326–330. [Google Scholar] [CrossRef]
  68. Fernandes, M.G.; Costa, J.; Reis, J.; Jacob, M.; Moura, C.; Machado, J.; Hespanhol, V. OA08.07 BRAF-V600E Advanced Lung Adenocarcinoma with Leptomeningeal (LM) Disease Treated with Vemurafenib. J. Thorac. Oncol. 2017, 12, S274–S275. [Google Scholar] [CrossRef]
  69. Falchook, G.S.; Long, G.V.; Kurzrock, R.; Kim, K.B.; Arkenau, T.H.; Brown, M.P.; Hamid, O.; Infante, J.R.; Millward, M.; Pavlick, A.C.; et al. Dabrafenib in patients with melanoma, untreated brain metastases, and other solid tumours: A phase 1 dose-escalation trial. Lancet 2012, 379, 1893–1901. [Google Scholar] [CrossRef]
  70. Desai, J.; Gan, H.; Barrow, C.; Jameson, M.; Atkinson, V.; Haydon, A.; Millward, M.; Begbie, S.; Brown, M.; Markman, B.; et al. Phase I, Open-Label, Dose-Escalation/Dose-Expansion Study of Lifirafenib (BGB-283), an RAF Family Kinase Inhibitor, in Patients with Solid Tumors. J. Clin. Oncol. 2020, 38, 2140–2150. [Google Scholar] [CrossRef]
  71. Poulikakos, P.I.; Persaud, Y.; Janakiraman, M.; Kong, X.; Ng, C.; Moriceau, G.; Shi, H.; Atefi, M.; Titz, B.; Gabay, M.T.; et al. RAF inhibitor resistance is mediated by dimerization of aberrantly spliced BRAF(V600E). Nature 2011, 480, 387–390. [Google Scholar] [CrossRef]
  72. Johannessen, C.M.; Boehm, J.S.; Kim, S.Y.; Thomas, S.R.; Wardwell, L.; Johnson, L.A.; Emery, C.M.; Stransky, N.; Cogdill, A.P.; Barretina, J.; et al. COT drives resistance to RAF inhibition through MAP kinase pathway reactivation. Nature 2010, 468, 968–972. [Google Scholar] [CrossRef]
  73. Chan, X.Y.; Singh, A.; Osman, N.; Piva, T.J. Role Played by Signalling Pathways in Overcoming BRAF Inhibitor Resistance in Melanoma. Int. J. Mol. Sci. 2017, 18, 1527. [Google Scholar] [CrossRef]
  74. Rudin, C.M.; Hong, K.; Streit, M. Molecular Characterization of Acquired Resistance to the BRAF Inhibitor Dabrafenib in a Patient with BRAF-Mutant Non–Small-Cell Lung Cancer. J. Thorac. Oncol. 2013, 8, e41–e42. [Google Scholar] [CrossRef]
  75. Schram, A.M.; Subbiah, V.; Sullivan, R.; Cosman, R.; Liu, J.; Sbar, E.I.; Hoang, T.; Chen, J.; Johnson, M.; Amoruccio, V.; et al. Abstract CT031: A first-in-human, phase 1a/1b, open-label, dose-escalation and expansion study to investigate the safety, pharmacokinetics, and antitumor activity of the RAF dimer inhibitor BGB-3245 in patients with advanced or refractory tumors. Cancer Res. 2023, 83, CT031. [Google Scholar] [CrossRef]
  76. Ng, P.; Han, Y.; Ogawa, L.S.; Schulz, R.; Yang, S.; Jewett, I.; Ishiyama, N.; Romashko, D.; Salomatov, A.; Thakur, S.; et al. Preclinical efficacy of BDTX-4933, a brain penetrant MasterKey inhibitor targeting oncogenic BRAF Class I/II/III mutations. Eur. J. Cancer 2022, 174, S86. [Google Scholar] [CrossRef]
  77. Chen, Y.K.; Kanouni, T.; Arnold, L.D.; Cox, J.M.; Gardiner, E.; Grandinetti, K.; Jiang, P.; Kaldor, S.W.; Lee, C.; Li, C.; et al. The Discovery of Exarafenib (KIN-2787): Overcoming the Challenges of Pan-RAF Kinase Inhibition. J. Med. Chem. 2024, 67, 1747–1757. [Google Scholar] [CrossRef]
  78. De La Fuente, M.I.; Ahnert, J.R.; Yaeger, R.; Tsai, F.Y.-C.; Janku, F.; Butowski, N.A.; Allen, C.E.; Ammakkanavar, N.R.; Taylor, J.W.; Michelson, G.; et al. Safety and efficacy of the novel BRAF inhibitor FORE8394 in patients with advanced solid and CNS tumors: Results from a phase 1/2a study. J. Clin. Oncol. 2023, 41, 3006. [Google Scholar] [CrossRef]
  79. Hainsworth, J.D.; Cebotaru, C.L.; Kanarev, V.; Ciuleanu, T.E.; Damyanov, D.; Stella, P.; Ganchev, H.; Pover, G.; Morris, C.; Tzekova, V. A Phase II, Open-Label, Randomized Study to Assess the Efficacy and Safety of AZD6244 (ARRY-142886) Versus Pemetrexed in Patients with Non-small Cell Lung Cancer Who Have Failed One or Two Prior Chemotherapeutic Regimens. J. Thorac. Oncol. 2010, 5, 1630–1636. [Google Scholar] [CrossRef]
  80. Haura, E.B.; Ricart, A.D.; Larson, T.G.; Stella, P.J.; Bazhenova, L.; Miller, V.A.; Cohen, R.B.; Eisenberg, P.D.; Selaru, P.; Wilner, K.D.; et al. A Phase II Study of PD-0325901, an Oral MEK Inhibitor, in Previously Treated Patients with Advanced Non–Small Cell Lung Cancer. Clin. Cancer Res. 2010, 16, 2450–2457. [Google Scholar] [CrossRef]
  81. Middleton, G.; Fletcher, P.; Popat, S.; Savage, J.; Summers, Y.; Greystoke, A.; Gilligan, D.; Cave, J.; O’rourke, N.; Brewster, A.; et al. The National Lung Matrix Trial of personalized therapy in lung cancer. Nature 2020, 583, 807–812. [Google Scholar] [CrossRef]
  82. Lopez-Chavez, A.; Thomas, A.; Rajan, A.; Raffeld, M.; Morrow, B.; Kelly, R.; Carter, C.A.; Guha, U.; Killian, K.; Lau, C.C.; 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, 1000–1007. [Google Scholar] [CrossRef]
  83. Chen, C.; Huang, C.; Xu, M.; Chen, Y.; Zhao, L.; Chen, Y.; Hu, Y. Abstract 475: Preclinical development of ABM-168, a novel MEK Inhibitor to treat cancer with brain tumors. Cancer Res. 2023, 83, 475. [Google Scholar] [CrossRef]
  84. Hall, B.; Travesa, A.; Yamamura, A.; Axel, A.; Kolitz, S.; Funt, J.; Fowler, K.D.; Nord, M.; Nair, P.; Barrett, S.; et al. Cyclic disruption of the mitogen-activated protein kinase (MAPK) pathway by the dual MEK inhibitor, IMM-6-415, enhances PD1 and CTLA4 checkpoint blockade in RAS mutant tumors. J. Immunother. Cancer 2022, 10, A469. [Google Scholar] [CrossRef]
  85. Long, G.V.; Stroyakovskiy, D.; Gogas, H.; Levchenko, E.; de Braud, F.; Larkin, J.; Garbe, C.; Jouary, T.; Hauschild, A.; Grob, J.-J.; et al. Dabrafenib and trametinib versus dabrafenib and placebo for Val600 BRAF-mutant melanoma: A multicentre, double-blind, phase 3 randomised controlled trial. Lancet 2015, 386, 444–451. [Google Scholar] [CrossRef]
  86. Planchard, D.; Smit, E.F.; Groen, H.J.M.; Mazieres, J.; Besse, B.; Helland, Å.; Giannone, V.; D’Amelio, A.M., Jr.; Zhang, P.; Mookerjee, B.; 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. [Google Scholar] [CrossRef]
  87. Planchard, D.; Besse, B.; Groen, H.J.; Hashemi, S.M.; Mazieres, J.; Kim, T.M.; Quoix, E.; Souquet, P.-J.; Barlesi, F.; Baik, C.; et al. Phase 2 Study of Dabrafenib Plus Trametinib in Patients with BRAF V600E-Mutant Metastatic NSCLC: Updated 5-Year Survival Rates and Genomic Analysis. J. Thorac. Oncol. 2021, 17, 103–115. [Google Scholar] [CrossRef]
  88. Savoia, P.; Fava, P.; Casoni, F.; Cremona, O. Targeting the ERK Signaling Pathway in Melanoma. Int. J. Mol. Sci. 2019, 20, 1483. [Google Scholar] [CrossRef]
  89. Kopetz, S.; Grothey, A.; Yaeger, R.; Van Cutsem, E.; Desai, J.; Yoshino, T.; Wasan, H.; Ciardiello, F.; Loupakis, F.; Hong, Y.S.; et al. Encorafenib, Binimetinib, and Cetuximab in BRAF V600E–Mutated Colorectal Cancer. N. Engl. J. Med. 2019, 381, 1632–1643. [Google Scholar] [CrossRef]
  90. Riely, G.J.; Ahn, M.J.; Felip, E.; Ramalingam, S.S.; Smit, E.F.; Tsao, A.S.; Alcasid, A.; Usari, T.; Wissel, P.S.; Wilner, K.D.; et al. Encorafenib plus binimetinib in patients with BRAFV600-mutant non-small cell lung cancer: Phase II PHAROS study design. Futur. Oncol. 2022, 18, 781–791. [Google Scholar] [CrossRef]
  91. Riely, G.J.; Smit, E.F.; Ahn, M.-J.; Felip, E.; Ramalingam, S.S.; Tsao, A.; Johnson, M.; Gelsomino, F.; Esper, R.; Nadal, E.; et al. Phase II, Open-Label Study of Encorafenib Plus Binimetinib in Patients with BRAFV600-Mutant Metastatic Non–Small-Cell Lung Cancer. J. Clin. Oncol. 2023, 41, 3700–3711. [Google Scholar] [CrossRef] [PubMed]
  92. Johnson, M.L.; Braiteh, F.; Grilley-Olson, J.E.; Chou, J.; Davda, J.; Forgie, A.; Li, R.; Jacobs, I.; Kazazi, F.; Hu-Lieskovan, S. Assessment of Subcutaneous vs Intravenous Administration of Anti–PD-1 Antibody PF-06801591 in Patients with Advanced Solid Tumors. JAMA Oncol. 2019, 5, 999–1007. [Google Scholar] [CrossRef] [PubMed]
  93. Boyer, M.; Patel, S.; Marron, T.; Pavlakis, N.; Parakh, S.; Gadgeel, S.; Shafique, M.; Hoyer, R.; Camidge, D.R.; Farber, C.; et al. A phase 1b/2 umbrella study of anti-PD-1 sasanlimab in combination with other therapies for patients with stage IIIB/IV non-small cell lung cancer (NSCLC): The LANDSCAPE 1011 trial in progress. J. Immunother. Cancer 2021, 9, A484. [Google Scholar] [CrossRef]
  94. Chen, S.H.; Gong, X.; Zhang, Y.; Van Horn, R.D.; Yin, T.; Huber, L.; Burke, T.F.; Manro, J.; Iversen, P.W.; Wu, W.; et al. RAF inhibitor LY3009120 sensitizes RAS or BRAF mutant cancer to CDK4/6 inhibition by abemaciclib via superior inhibition of phospho-RB and suppression of cyclin D1. Oncogene 2018, 37, 821–832. [Google Scholar] [CrossRef] [PubMed]
  95. Verastem Oncology Granted Fast Track Designation for Combination of Avutometinib and Sotorasib for the Treatment of KRAS G12C-Mutant Non-Small Cell Lung Cancer (NSCLC). News Release. Verastem Oncology. 18 January 2024. Available online: https://tinyurl.com/vmn25kcy (accessed on 12 April 2024).
  96. Govindan, R.; Awad, M.M.; Gadgeel, S.M.; Pachter, J.A.; Patrick, G.; Denis, L.J. A phase 1/2 study of VS-6766 (RAF/MEK clamp) in combination with sotorasib (G12C inhibitor) in patients with KRAS G12C mutant non–small cell lung cancer (NSCLC) (RAMP 203). J. Clin. Oncol. 2022, 40 (Suppl. S16), TPS914. [Google Scholar]
  97. Minchom, A.R.; Perez, V.S.; Morton, C.; ThManickavasagar, T.; Nintos, G.; Lai-Kwon, J.E.; Guo, C.; Tunariu, N.; Parker, T.; Prout, T.; et al. Phase I trial of the RAF/MEK clamp VS-6766 in combination with everolimus using an intermittent schedule with expansion in NSCLC across multiple KRAS variants. J. Clin. Oncol. 2022, 40 (Suppl. S16), 9018. [Google Scholar] [CrossRef]
  98. Oxnard, G.R.; Yang, J.C.; Yu, H.; Kim, S.W.; Saka, H.; Horn, L.; Goto, K.; Ohe, Y.; Mann, H.; Thress, K.S.; et al. TATTON: A multi-arm, phase Ib trial of osimertinib combined with selumetinib, savolitinib, or durvalumab in EGFR-mutant lung cancer. Ann. Oncol. 2020, 31, 507–516. [Google Scholar] [CrossRef] [PubMed]
  99. Lito, P.; Pratilas, C.A.; Joseph, E.W.; Tadi, M.; Halilovic, E.; Zubrowski, M.; Huang, A.; Wong, W.L.; Callahan, M.K.; Merghoub, T.; et al. Relief of Profound Feedback Inhibition of Mitogenic Signaling by RAF Inhibitors Attenuates Their Activity in BRAFV600E Melanomas. Cancer Cell 2012, 22, 668–682. [Google Scholar] [CrossRef] [PubMed]
  100. Corcoran, R.B.; Ebi, H.; Turke, A.B.; Coffee, E.M.; Nishino, M.; Cogdill, A.P.; Brown, R.D.; Della Pelle, P.; Dias-Santagata, D.; Hung, K.E.; et al. EGFR-Mediated Reactivation of MAPK Signaling Contributes to Insensitivity of BRAF-Mutant Colorectal Cancers to RAF Inhibition with Vemurafenib. Cancer Discov. 2012, 2, 227–235. [Google Scholar] [CrossRef]
  101. Villanueva, J.; Vultur, A.; Herlyn, M. Resistance to BRAF Inhibitors: Unraveling Mechanisms and Future Treatment Options. Cancer Res. 2011, 71, 7137–7140. [Google Scholar] [CrossRef]
  102. Johnson, D.B.; Menzies, A.M.; Zimmer, L.; Eroglu, Z.; Ye, F.; Zhao, S.; Rizos, H.; Sucker, A.; Scolyer, R.A.; Gutzmer, R.; et al. Acquired BRAF inhibitor resistance: A multicenter meta-analysis of the spectrum and frequencies, clinical behaviour, and phenotypic associations of resistance mechanisms. Eur. J. Cancer 2015, 51, 2792–2799. [Google Scholar] [CrossRef] [PubMed]
  103. Van Allen, E.M.; Wagle, N.; Sucker, A.; Treacy, D.J.; Johannessen, C.M.; Goetz, E.M.; Place, C.S.; Taylor-Weiner, A.; Whittaker, S.; Kryukov, G.V.; et al. The Genetic Landscape of Clinical Resistance to RAF Inhibition in Metastatic Melanoma. Cancer Discov. 2014, 4, 94–109. [Google Scholar] [CrossRef] [PubMed]
  104. Shi, H.; Hong, A.; Kong, X.; Koya, R.C.; Song, C.; Moriceau, G.; Hugo, W.; Yu, C.C.; Ng, C.; Chodon, T.; et al. A Novel AKT1 Mutant Amplifies an Adaptive Melanoma Response to BRAF Inhibition. Cancer Discov. 2014, 4, 69–79. [Google Scholar] [CrossRef]
  105. Shi, H.; Hugo, W.; Kong, X.; Hong, A.; Koya, R.C.; Moriceau, G.; Chodon, T.; Guo, R.; Johnson, D.B.; Dahlman, K.B.; et al. Acquired Resistance and Clonal Evolution in Melanoma during BRAF Inhibitor Therapy. Cancer Discov. 2014, 4, 80–93. [Google Scholar] [CrossRef]
  106. Niemantsverdriet, M.; Schuuring, E.; ter Elst, A.; van der Wekken, A.J.; van Kempen, L.C.; Berg, A.v.D.; Groen, H.J. KRAS Mutation as a Resistance Mechanism to BRAF/MEK Inhibition in NSCLC. J. Thorac. Oncol. 2018, 13, e249–e251. [Google Scholar] [CrossRef] [PubMed]
  107. Abravanel, D.L.; Nishino, M.; Sholl, L.M.; Ambrogio, C.; Awad, M.M. An Acquired NRAS Q61K Mutation in BRAF V600E-Mutant Lung Adenocarcinoma Resistant to Dabrafenib Plus Trametinib. J. Thorac. Oncol. 2018, 13, e131–e133. [Google Scholar] [CrossRef]
  108. Facchinetti, F.; Lacroix, L.; Mezquita, L.; Scoazec, J.-Y.; Loriot, Y.; Tselikas, L.; Gazzah, A.; Rouleau, E.; Adam, J.; Michiels, S.; et al. Molecular mechanisms of resistance to BRAF and MEK inhibitors in BRAFV600E non–small cell lung cancer. Eur. J. Cancer 2020, 132, 211–223. [Google Scholar] [CrossRef]
  109. Wang, V.E.; Xue, J.Y.; Frederick, D.T.; Cao, Y.; Lin, E.; Wilson, C.; Urisman, A.; Carbone, D.P.; Flaherty, K.T.; Bernards, R.; et al. Adaptive Resistance to Dual BRAF/MEK Inhibition in BRAF-Driven Tumors through Autocrine FGFR Pathway Activation. Clin. Cancer Res. 2019, 25, 7202–7217. [Google Scholar] [CrossRef]
  110. Manchado, E.; Weissmueller, S.; Morris, J.P.; Chen, C.C.; Wullenkord, R.; Lujambio, A.; de Stanchina, E.; Poirier, J.T.; Gainor, J.F.; Corcoran, R.B.; et al. A combinatorial strategy for treating KRAS-mutant lung cancer. Nature 2016, 534, 647–651. [Google Scholar] [CrossRef]
  111. Hirai, N.; Hatanaka, Y.; Hatanaka, K.C.; Uno, Y.; Chiba, S.I.; Umekage, Y.; Minami, Y.; Okumura, S.; Ohsaki, Y.; Sasaki, T. Cyclin-dependent kinase 4 upregulation mediates acquired resistance of dabrafenib plus trametinib in BRAF V600E-mutated lung cancer. Transl. Lung Cancer Res. 2021, 10, 3737–3744. [Google Scholar] [CrossRef]
  112. Marusiak, A.A.; Edwards, Z.C.; Hugo, W.; Trotter, E.W.; Girotti, M.R.; Stephenson, N.L.; Kong, X.; Gartside, M.G.; Fawdar, S.; Hudson, A.; et al. Mixed lineage kinases activate MEK independently of RAF to mediate resistance to RAF inhibitors. Nat. Commun. 2014, 5, 3901. [Google Scholar] [CrossRef]
  113. Lionarons, D.A.; Hancock, D.C.; Rana, S.; East, P.; Moore, C.; Murillo, M.M.; Carvalho, J.; Spencer-Dene, B.; Herbert, E.; Stamp, G.; et al. RAC1P29S Induces a Mesenchymal Phenotypic Switch via Serum Response Factor to Promote Melanoma Development and Therapy Resistance. Cancer Cell 2019, 36, 68–83.e9. [Google Scholar] [CrossRef] [PubMed]
  114. Hanrahan, A.J.; Sylvester, B.E.; Chang, M.T.; Elzein, A.; Gao, J.; Han, W.; Liu, Y.; Xu, D.; Gao, S.P.; Gorelick, A.N.; et al. Leveraging Systematic Functional Analysis to Benchmark an In Silico Framework Distinguishes Driver from Passenger MEK Mutants in Cancer. Cancer Res. 2020, 80, 4233–4243. [Google Scholar] [CrossRef] [PubMed]
  115. Gao, Y.; Chang, M.T.; McKay, D.; Na, N.; Zhou, B.; Yaeger, R.; Torres, N.M.; Muniz, K.; Drosten, M.; Barbacid, M.; et al. Allele-Specific Mechanisms of Activation of MEK1 Mutants Determine Their Properties. Cancer Discov. 2018, 8, 648–661. [Google Scholar] [CrossRef] [PubMed]
  116. Nathanson, K.L.; Martin, A.M.; Wubbenhorst, B.; Greshock, J.; Letrero, R.; D’Andrea, K.; O’Day, S.; Infante, J.R.; Falchook, G.S.; Arkenau, H.T.; et al. Tumor genetic analyses of patients with metastatic melanoma treated with the BRAF inhibitor dabrafenib (GSK2118436). Clin. Cancer Res. 2013, 19, 4868–4878. [Google Scholar] [CrossRef] [PubMed]
  117. Smalley, K.S.; Lioni, M.; Dalla Palma, M.; Xiao, M.; Desai, B.; Egyhazi, S.; Hansson, J.; Wu, H.; King, A.J.; Van Belle, P.; et al. Increased cyclin D1 expression can mediate BRAF inhibitor resistance in BRAF V600E-mutated melanomas. Mol. Cancer Ther. 2008, 7, 2876–2883. [Google Scholar] [CrossRef] [PubMed]
  118. Nassar, K.W.; Hintzsche, J.D.; Bagby, S.M.; Espinoza, V.; Langouët-Astrié, C.; Amato, C.M.; Chimed, T.S.; Fujita, M.; Robinson, W.; Tan, A.C.; et al. Targeting CDK4/6 Represents a Therapeutic Vulnerability in Acquired BRAF/MEK Inhibitor-Resistant Melanoma. Mol. Cancer Ther. 2021, 20, 2049–2060. [Google Scholar] [CrossRef] [PubMed]
  119. Gelsomino, F.; Di Federico, A.; Tardio, M.L.; Grilli, G.; D’errico, A.; Ardizzoni, A.; Salvagni, S. Drug-induced colitis on BRAF and MEK inhibitors for BRAF V600E-mutated non-small cell lung cancer: A case report. Investig. New Drugs 2021, 40, 190–193. [Google Scholar] [CrossRef]
  120. Han, J.; Liu, Y.; Yang, S.; Wu, X.; Li, H.; Wang, Q. MEK inhibitors for the treatment of non-small cell lung cancer. J. Hematol. Oncol. 2021, 14, 1. [Google Scholar] [CrossRef]
  121. Garutti, M.; Bergnach, M.; Polesel, J.; Palmero, L.; Pizzichetta, M.A.; Puglisi, F. BRAF and MEK Inhibitors and Their Toxicities: A Meta-Analysis. Cancers 2022, 15, 141. [Google Scholar] [CrossRef]
  122. Ribas, A.; Algazi, A.; Ascierto, P.A.; Butler, M.O.; Chandra, S.; Gordon, M.; Hernandez-Aya, L.; Lawrence, D.; Lutzky, J.; Miller, W.H., Jr.; et al. PD-L1 blockade in combination with inhibition of MAPK oncogenic signaling in patients with advanced melanoma. Nat. Commun. 2020, 11, 6262. [Google Scholar] [CrossRef] [PubMed]
  123. Gogas, H.; Dréno, B.; Larkin, J.; Demidov, L.; Stroyakovskiy, D.; Eroglu, Z.; Francesco Ferrucci, P.; Pigozzo, J.; Rutkowski, P.; Mackiewicz, J.; et al. Cobimetinib plus atezolizumab in BRAFV600 wild-type melanoma: Primary results from the randomized phase III IMspire170 study. Ann. Oncol. 2021, 32, 384–394. [Google Scholar] [CrossRef] [PubMed]
  124. Gutzmer, R.; Stroyakovskiy, D.; Gogas, H.; Robert, C.; Lewis, K.; Protsenko, S.; Pereira, R.P.; Eigentler, T.; Rutkowski, P.; Demidov, L.; et al. Atezolizumab, vemurafenib, and cobimetinib as first-line treatment for unresectable advanced BRAFV600 mutation-positive melanoma (IMspire150): Primary analysis of the randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2020, 395, 1835–1844. [Google Scholar] [CrossRef] [PubMed]
  125. Della Corte, C.M.; Barra, G.; Ciaramella, V.; Di Liello, R.; Vicidomini, G.; Zappavigna, S.; Luce, A.; Abate, M.; Fiorelli, A.; Caraglia, M.; et al. Antitumor activity of dual blockade of PD-L1 and MEK in NSCLC patients derived three-dimensional spheroid cultures. J. Exp. Clin. Cancer Res. 2019, 38, 253. [Google Scholar] [CrossRef] [PubMed]
  126. Lee, J.W.; Zhang, Y.; Eoh, K.J.; Sharma, R.; Sanmamed, M.F.; Wu, J.; Choi, J.; Park, H.S.; Iwasaki, A.; Kaftan, E.; et al. The Combination of MEK Inhibitor with Immunomodulatory Antibodies Targeting Programmed Death 1 and Programmed Death Ligand 1 Results in Prolonged Survival in Kras/p53-Driven Lung Cancer. J. Thorac. Oncol. 2019, 14, 1046–1060. [Google Scholar] [CrossRef] [PubMed]
  127. Coma, S.; Arcas, M.M.; Downward, J.; Pachter, J.A. The RAF/MEK clamp avutometinib (VS-6766) induces an immunogenic tumor microenvironment and potentiates the efficacy of anti-PD-1. Cancer Res. 2023, 83 (Suppl. S7), 4155. [Google Scholar] [CrossRef]
  128. Gaudreau, P.O.; Lee, J.J.; Heymach, J.V.; Gibbons, D.L. Phase I/II Trial of Immunotherapy with Durvalumab and Tremelimumab with Continuous or Intermittent MEK Inhibitor Selumetinib in NSCLC: Early Trial Report. Clin. Lung Cancer 2020, 21, 384–388. [Google Scholar] [CrossRef] [PubMed]
  129. Sumimoto, H.; Imabayashi, F.; Iwata, T.; Kawakami, Y. The BRAF–MAPK signaling pathway is essential for cancer-immune evasion in human melanoma cells. J. Exp. Med. 2006, 203, 1651–1656. [Google Scholar] [CrossRef] [PubMed]
  130. Wilmott, J.S.; Long, G.V.; Howle, J.R.; Haydu, L.E.; Sharma, R.N.; Thompson, J.F.; Kefford, R.F.; Hersey, P.; Scolyer, R.A. Selective BRAF Inhibitors Induce Marked T-cell Infiltration into Human Metastatic Melanoma. Clin. Cancer Res. 2012, 18, 1386–1394. [Google Scholar] [CrossRef]
  131. Frederick, D.T.; Piris, A.; Cogdill, A.P.; Cooper, Z.A.; Lezcano, C.; Ferrone, C.R.; Mitra, D.; Boni, A.; Newton, L.P.; Liu, C.; et al. BRAF Inhibition Is Associated with Enhanced Melanoma Antigen Expression and a More Favorable Tumor Microenvironment in Patients with Metastatic Melanoma. Clin. Cancer Res. 2013, 19, 1225–1231. [Google Scholar] [CrossRef]
  132. Boni, A.; Cogdill, A.P.; Dang, P.; Udayakumar, D.; Njauw, C.N.J.; Sloss, C.M.; Ferrone, C.R.; Flaherty, K.T.; Lawrence, D.P.; Fisher, D.E.; et al. Selective BRAFV600E Inhibition Enhances T-Cell Recognition of Melanoma without Affecting Lymphocyte Function. Cancer Res. 2010, 70, 5213–5219. [Google Scholar] [CrossRef] [PubMed]
  133. Poon, E.; Mullins, S.; Watkins, A.; Williams, G.S.; Koopmann, J.O.; Di Genova, G.; Cumberbatch, M.; Veldman-Jones, M.; Grosskurth, S.E.; Sah, V.; et al. The MEK inhibitor selumetinib complements CTLA-4 blockade by reprogramming the tumor immune microenvironment. J. Immunother. Cancer 2017, 5, 63. [Google Scholar] [CrossRef] [PubMed]
  134. Choi, H.; Deng, J.; Li, S.; Silk, T.; Dong, L.; Brea, E.J.; Houghton, S.; Redmond, D.; Zhong, H.; Boiarsky, J.; et al. Pulsatile MEK Inhibition Improves Anti-tumor Immunity and T Cell Function in Murine Kras Mutant Lung Cancer. Cell Rep. 2019, 27, 806–819.e5. [Google Scholar] [CrossRef] [PubMed]
  135. Niu, X.; Sun, Y.; Planchard, D.; Chiu, L.; Bai, J.; Ai, X.; Lu, S. Durable Response to the Combination of Atezolizumab With Platinum-Based Chemotherapy in an Untreated Non-Smoking Lung Adenocarcinoma Patient with BRAF V600E Mutation: A Case Report. Front. Oncol. 2021, 11, 634920. [Google Scholar] [CrossRef]
  136. Hellmann, M.D.; Kim, T.W.; Lee, C.; Goh, B.C.; Miller, W.; Oh, D.Y.; Jamal, R.; Chee, C.E.; Chow, L.; Gainor, J.; et al. Phase Ib study of atezolizumab combined with cobimetinib in patients with solid tumors. Ann. Oncol. 2019, 30, 1134–1142. [Google Scholar] [CrossRef] [PubMed]
  137. Okimoto, R.A.; Lin, L.; Olivas, V.; Chan, E.; Markegard, E.; Rymar, A.; Neel, D.; Chen, X.; Hemmati, G.; Bollag, G.; et al. Preclinical efficacy of a RAF inhibitor that evades paradoxical MAPK pathway activation in protein kinase BRAF-mutant lung cancer. Proc. Natl. Acad. Sci. USA 2016, 113, 13456–13461. [Google Scholar] [CrossRef]
  138. Cao, X.X.; Wu, Y.; Liu, P.; Ding, T.; Ye, H.; Cai, Z.; Zhang, Y.; Ma, J.; Xu, Y.; Fu, G.; et al. 329MO HLX208, a novel BRAF V600E inhibitor, in adult patients with Langerhans cell histiocytosis and/or Erdheim-Chester disease harbouring BRAF V600E mutation. Ann. Oncol. 2023, 34, S1599. [Google Scholar] [CrossRef]
  139. Cao, X.X.; Wu, Y.; Liu, P.; Ding, T.; Ye, H.; Cai, Z.; Zhang, Y.; Hu, C.; Hou, X.; Yang, G.; et al. A phase 2 study of HLX208, a BRAFV600E inhibitor, in adult patients with Langerhans cell histiocytosis and/or Erdheim-Chester disease harboring BRAFV600E mutation. J. Clin. Oncol. 2023, 41, 7574. [Google Scholar] [CrossRef]
  140. Arozarena, I.; Wellbrock, C. Overcoming resistance to BRAF inhibitors. Ann. Transl. Med. 2017, 5, 387. [Google Scholar] [CrossRef]
  141. Algazi, A.P.; Othus, M.; Daud, A.I.; Lo, R.S.; Mehnert, J.M.; Truong, T.G.; Conry, R.; Kendra, K.; Doolittle, G.C.; Clark, J.I.; et al. Continuous versus intermittent BRAF and MEK inhibition in patients with BRAF-mutated melanoma: A randomized phase 2 trial. Nat. Med. 2020, 26, 1564–1568. [Google Scholar] [CrossRef]
  142. Das Thakur, M.; Salangsang, F.; Landman, A.S.; Sellers, W.R.; Pryer, N.K.; Levesque, M.P.; Dummer, R.; McMahon, M.; Stuart, D.D. Modelling vemurafenib resistance in melanoma reveals a strategy to forestall drug resistance. Nature 2013, 494, 251–255. [Google Scholar] [CrossRef] [PubMed]
  143. Kashizaki, F.; Tanaka, A.; Hattori, S.; Sugimoto, S. Dabrafenib–trametinib combination therapy re-challenge in advanced BRAFV600E-mutant non–small-cell lung cancer. Eur. J. Cancer 2020, 143, 31–32. [Google Scholar] [CrossRef] [PubMed]
  144. Bhagwat, S.V.; McMillen, W.T.; Cai, S.; Zhao, B.; Whitesell, M.; Shen, W.; Kindler, L.; Flack, R.S.; Wu, W.; Anderson, B.; et al. ERK Inhibitor LY3214996 Targets ERK Pathway–Driven Cancers: A Therapeutic Approach Toward Precision Medicine. Mol. Cancer Ther. 2020, 19, 325–336. [Google Scholar] [CrossRef] [PubMed]
  145. Bardia, A.; Gounder, M.; Rodon, J.; Janku, F.; Lolkema, M.P.; Stephenson, J.J.; Bedard, P.L.; Schuler, M.; Sessa, C.; LoRusso, P.; et al. Phase Ib Study of Combination Therapy with MEK Inhibitor Binimetinib and Phosphatidylinositol 3-Kinase Inhibitor Buparlisib in Patients with Advanced Solid Tumors with RAS/RAF Alterations. The Oncologist 2019, 25, e160–e169. [Google Scholar] [CrossRef]
  146. Ojha, R.; Leli, N.M.; Onorati, A.; Piao, S.; Verginadis, I.I.; Tameire, F.; Rebecca, V.W.; Chude, C.I.; Murugan, S.; Fennelly, C.; et al. ER Translocation of the MAPK Pathway Drives Therapy Resistance in BRAF-Mutant Melanoma. Cancer Discov. 2019, 9, 396–415. [Google Scholar] [CrossRef]
  147. Mehnert, J.M.; Mitchell, T.C.; Huang, A.C.; Aleman, T.S.; Kim, B.J.; Schuchter, L.M.; Linette, G.P.; Karakousis, G.C.; Mitnick, S.; Giles, L.; et al. BAMM (BRAF Autophagy and MEK Inhibition in Melanoma): A Phase I/II Trial of Dabrafenib, Trametinib, and Hydroxychloroquine in Advanced BRAFV600-mutant Melanoma. Clin. Cancer Res. 2022, 28, 1098–1106. [Google Scholar] [CrossRef]
  148. Ma, X.-H.; Piao, S.-F.; Dey, S.; Mcafee, Q.; Karakousis, G.; Villanueva, J.; Hart, L.S.; Levi, S.; Hu, J.; Zhang, G.; et al. Targeting ER stress–induced autophagy overcomes BRAF inhibitor resistance in melanoma. J. Clin. Investig. 2014, 124, 1406–1417. [Google Scholar] [CrossRef]
  149. Tolcher, A.W.; Hong, D.S.; Vandross, A.L.; Psoinos, C.M.; Brennan, D.M.; Sherman, M.L.; Ruiz-Soto, R.; Reu, F.J.; Weekes, C.D. A phase 1/2 study of DCC-3116 as a single agent and in combination with trametinib in patients with advanced or metastatic solid tumors with RAS or RAF mutations. J. Clin. Oncol. 2022, 40, TPS3178. [Google Scholar] [CrossRef]
  150. Gao, W.; Wang, X.; Zhou, Y.; Wang, X.; Yu, Y. Autophagy, ferroptosis, pyroptosis, and necroptosis in tumor immunotherapy. Signal Transduct. Target. Ther. 2022, 7, 196. [Google Scholar] [CrossRef]
  151. Zhang, Z.; Zhang, Y.; Lieberman, J. Lighting a Fire: Can We Harness Pyroptosis to Ignite Antitumor Immunity? Cancer Immunol. Res. 2021, 9, 2–7. [Google Scholar] [CrossRef]
  152. Erkes, D.A.; Cai, W.; Sanchez, I.M.; Purwin, T.J.; Rogers, C.; Field, C.O.; Berger, A.C.; Hartsough, E.J.; Rodeck, U.; Alnemri, E.S.; et al. Mutant BRAF and MEK Inhibitors Regulate the Tumor Immune Microenvironment via Pyroptosis. Cancer Discov. 2020, 10, 254–269. [Google Scholar] [CrossRef] [PubMed]
  153. Kano, H.; Ichihara, E.; Watanabe, H.; Nishii, K.; Ando, C.; Nakasuka, T.; Ninomiya, K.; Kato, Y.; Kubo, T.; Rai, K.; et al. SHP2 Inhibition Enhances the Effects of Tyrosine Kinase Inhibitors in Preclinical Models of Treatment-naïve ALK-, ROS1-, or EGFR-altered Non–small Cell Lung Cancer. Mol. Cancer Ther. 2021, 20, 1653–1662. [Google Scholar] [CrossRef] [PubMed]
  154. Mainardi, S.; Mulero-Sánchez, A.; Prahallad, A.; Germano, G.; Bosma, A.; Krimpenfort, P.; Lieftink, C.; Steinberg, J.D.; de Wit, N.; Gonçalves-Ribeiro, S.; et al. SHP2 is required for growth of KRAS-mutant non-small-cell lung cancer in vivo. Nat. Med. 2018, 24, 961–967. [Google Scholar] [CrossRef]
  155. Fedele, C.; Ran, H.; Diskin, B.; Wei, W.; Jen, J.; Geer, M.J.; Araki, K.; Ozerdem, U.; Simeone, D.M.; Miller, G.; et al. SHP2 Inhibition Prevents Adaptive Resistance to MEK Inhibitors in Multiple Cancer Models. Cancer Discov. 2018, 8, 1237–1249. [Google Scholar] [CrossRef] [PubMed]
  156. Ahmed, T.A.; Adamopoulos, C.; Karoulia, Z.; Wu, X.; Sachidanandam, R.; Aaronson, S.A.; Poulikakos, P.I. SHP2 Drives Adaptive Resistance to ERK Signaling Inhibition in Molecularly Defined Subsets of ERK-Dependent Tumors. Cell Rep. 2019, 26, 65–78.e5. [Google Scholar] [CrossRef] [PubMed]
  157. Nichols, R.J.; Haderk, F.; Stahlhut, C.; Schulze, C.J.; Hemmati, G.; Wildes, D.; Tzitzilonis, C.; Mordec, K.; Marquez, A.; Romero, J.; et al. RAS nucleotide cycling underlies the SHP2 phosphatase dependence of mutant BRAF-, NF1- and RAS-driven cancers. Nat. Cell. Biol. 2018, 20, 1064–1073. [Google Scholar] [CrossRef] [PubMed]
  158. Wang, J.; Pollard, K.; Allen, A.N.; Tomar, T.; Pijnenburg, D.; Yao, Z.; Rodriguez, F.J.; Pratilas, C.A. Combined Inhibition of SHP2 and MEK Is Effective in Models of NF1-Deficient Malignant Peripheral Nerve Sheath Tumors. Cancer Res. 2020, 80, 5367–5379. [Google Scholar] [CrossRef]
  159. Drilon, A.E.; Johnson, M.L.; Gadgeel, S.M.; Nepert, D.; Feng, G.; Golmakani, M.; Reddy, M.; Harney, A.; Oliver, C.; Mills, B.; et al. A first-in-human, phase 1 study of the SHP2 inhibitor PF-07284892 as monotherapy and in combination with different targeted therapies in oncogene-driven, treatment-resistant solid tumors. J. Clin. Oncol. 2023, 41, 3020. [Google Scholar] [CrossRef]
  160. Drilon, A.; Sharma, M.R.; Johnson, M.L.; Yap, T.A.; Gadgeel, S.; Nepert, D.; Feng, G.; Reddy, M.B.; Harney, A.S.; Elsayed, M.; et al. SHP2 Inhibition Sensitizes Diverse Oncogene-Addicted Solid Tumors to Re-treatment with Targeted Therapy. Cancer Discov. 2023, 13, 1789–1801. [Google Scholar] [CrossRef]
  161. Hanna, N.H.; Robinson, A.G.; Temin, S.; Baker, S., Jr.; Brahmer, J.R.; Ellis, P.M.; Gaspar, L.E.; Haddad, R.Y.; Hesketh, P.J.; Jain, D.; et al. Therapy for Stage IV Non–Small-Cell Lung Cancer with Driver Alterations: ASCO and OH (CCO) Joint Guideline Update. J. Clin. Oncol. 2021, 39, 1040–1091. [Google Scholar] [CrossRef]
  162. de Braud, F.; Dooms, C.; Heist, R.S.; Lebbe, C.; Wermke, M.; Gazzah, A.; Schadendorf, D.; Rutkowski, P.; Wolf, J.; Ascierto, P.A.; et al. Initial Evidence for the Efficacy of Naporafenib in Combination with Trametinib in NRAS-Mutant Melanoma: Results from the Expansion Arm of a Phase Ib, Open-Label Study. J. Clin. Oncol. 2023, 41, 2651–2660. [Google Scholar] [CrossRef] [PubMed]
  163. Kim, T.W.; Lee, J.; Kim, T.M.; Shin, S.J.; Han, S.W.; Kim, J.S.; Kim, Y.J.; Yoo, C.; Hong, Y.S.; Kim, J.W.; et al. 529P A phase Ib trial of belvarafenib in combination with cobimetinib in patients (pts) with RAS- or RAF- mutated (m) solid tumors: Updated safety data and indication-specific efficacy results. Ann. Oncol. 2021, 32 (Suppl. S5), S595. [Google Scholar] [CrossRef]
  164. Ettinger, D.S.; Wood, D.E.; Aisner, D.L.; Akerley, W.; Bauman, J.R.; Bharat, A.; Bruno, D.S.; Chang, J.Y.; Chirieac, L.R.; D’Amico, T.A.; et al. Non-Small Cell Lung Cancer, Version 3.2022, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Canc. Netw. 2022, 20, 497–530. [Google Scholar] [CrossRef] [PubMed]
  165. Parekh, P.R.; Botting, G.M.; Thurber, D.B.; Boruszczak, M.; Murphy, W.; Bertenshaw, G.P. Predictive biomarkers for response to trametinib in non-small cell lung cancer. Tumour Biol. 2022, 44, 249–267. [Google Scholar] [CrossRef] [PubMed]
  166. Rustgi, N.; Maria, A.; Toumbacaris, N.; Zhao, H.; Kargus, K.; Bryant, M.; Waksmundzki, A.; Aricescu, I.; Lefkowitz, R.A.; Li, B.T.; et al. Combined RAF and MEK Inhibition to Treat Activated Non-V600 BRAF-Altered Advanced Cancers. Oncologist 2024, 29, 15–24. [Google Scholar] [CrossRef]
  167. Cords, L.; Engler, S.; Haberecker, M.; Rüschoff, J.H.; Moch, H.; de Souza, N.; Bodenmiller, B. Cancer-associated fibroblast phenotypes are associated with patient outcome in non-small cell lung cancer. Cancer Cell 2024, 42, 396–412.e5. [Google Scholar] [CrossRef]
Figure 1. The RTK/RAS/MAPK pathway, its key components and mutations in lung cancer and the different categories of RAF inhibitors based on their distinct structural properties. RTK, receptor tyrosine kinase; SHP2, Src homology 2 domain-containing phosphatase 2; SOS1, son of sevenless homolog; KRAS, Kirsten rat sarcoma viral oncogene homolog; GDP, guanosine diphosphate; GTP, guanosine triphosphate; BRAF, v-RAF murine sarcoma viral oncogene homolog B; MEK, mitogen-activated protein-kinase kinase; ERK, extracellular signal-regulated kinase; AP-1, activator protein-1; c-MYC, cellular Myelocytomatosis oncogene; ELK-1, ETS like-1; NF1, neurofibromin 1; EGFR, epidermal growth factor receptor; ALK, anaplastic lymphoma kinase; ROS1, c-ros oncogene-1; MET, hepatocyte growth factor receptor; HER2, human epidermal growth factor receptor 2; RET, rearranged during transfection; NTRK, neurotrophic tropomyosin receptor kinase. This figure was created using the tools provided by BioRender.com (accessed on 10 April 2024).
Figure 1. The RTK/RAS/MAPK pathway, its key components and mutations in lung cancer and the different categories of RAF inhibitors based on their distinct structural properties. RTK, receptor tyrosine kinase; SHP2, Src homology 2 domain-containing phosphatase 2; SOS1, son of sevenless homolog; KRAS, Kirsten rat sarcoma viral oncogene homolog; GDP, guanosine diphosphate; GTP, guanosine triphosphate; BRAF, v-RAF murine sarcoma viral oncogene homolog B; MEK, mitogen-activated protein-kinase kinase; ERK, extracellular signal-regulated kinase; AP-1, activator protein-1; c-MYC, cellular Myelocytomatosis oncogene; ELK-1, ETS like-1; NF1, neurofibromin 1; EGFR, epidermal growth factor receptor; ALK, anaplastic lymphoma kinase; ROS1, c-ros oncogene-1; MET, hepatocyte growth factor receptor; HER2, human epidermal growth factor receptor 2; RET, rearranged during transfection; NTRK, neurotrophic tropomyosin receptor kinase. This figure was created using the tools provided by BioRender.com (accessed on 10 April 2024).
Ijms 25 04633 g001
Figure 2. BRAF and MEK inhibitors under clinical evaluation in combination, or not, with various targeted therapies for lung cancer. RTK, receptor tyrosine kinase; SHP2, Src homology 2 domain-containing phosphatase 2; SOS1, son of sevenless homolog; KRAS, Kirsten rat sarcoma viral oncogene homolog; GDP, guanosine diphosphate; GTP, guanosine triphosphate; RAF, rapidly accelerated fibrosarcoma; MEK, mitogen-activated protein-kinase kinase; ERK, extracellular signal-regulated kinase; PI3K, phosphatidylinositol 3-kinase; PIP2/3, phosphatidylinositol 2/3; PTEN, phosphatase and tensin homolog; AKT, protein kinase B; mTOR, mechanistic target of rapamycin; EGFR, epidermal growth factor receptor; HER2, human epidermal growth factor receptor 2; ROS1, c-ros oncogene-1; MET, hepatocyte growth factor receptor; ALK, anaplastic lymphoma kinase; KIT, tyrosine-protein kinase kit; PDGFR, platelet-derived growth factor receptor; TRK, tropomyosin receptor kinase; VEGFR, vascular endothelial growth factor receptor; RET, rearranged during transfection; FGFR, fibroblast growth factor receptor; SMO, smoothened; PD-L1, programmed death-ligand 1; PD-1, programmed cell death protein 1; CTLA-4, cytotoxic T-lymphocyte-associated protein 4; LAG-3, lymphocyte-activation gene 3; TIGIT, T cell immunoreceptor with Ig and ITIM domains; FAK, focal adhesion kinase; SRC, proto-oncogene tyrosine-protein kinase; ULK1/2, unc-51-like kinase 1/2; CDK4/6, cyclin-dependent kinase 4/6; WEE-1, Wee1-like protein kinase; ATR, ataxia telangiectasia and Rad3-related protein. This figure was created using the tools provided by BioRender.com (accessed on 10 April 2024).
Figure 2. BRAF and MEK inhibitors under clinical evaluation in combination, or not, with various targeted therapies for lung cancer. RTK, receptor tyrosine kinase; SHP2, Src homology 2 domain-containing phosphatase 2; SOS1, son of sevenless homolog; KRAS, Kirsten rat sarcoma viral oncogene homolog; GDP, guanosine diphosphate; GTP, guanosine triphosphate; RAF, rapidly accelerated fibrosarcoma; MEK, mitogen-activated protein-kinase kinase; ERK, extracellular signal-regulated kinase; PI3K, phosphatidylinositol 3-kinase; PIP2/3, phosphatidylinositol 2/3; PTEN, phosphatase and tensin homolog; AKT, protein kinase B; mTOR, mechanistic target of rapamycin; EGFR, epidermal growth factor receptor; HER2, human epidermal growth factor receptor 2; ROS1, c-ros oncogene-1; MET, hepatocyte growth factor receptor; ALK, anaplastic lymphoma kinase; KIT, tyrosine-protein kinase kit; PDGFR, platelet-derived growth factor receptor; TRK, tropomyosin receptor kinase; VEGFR, vascular endothelial growth factor receptor; RET, rearranged during transfection; FGFR, fibroblast growth factor receptor; SMO, smoothened; PD-L1, programmed death-ligand 1; PD-1, programmed cell death protein 1; CTLA-4, cytotoxic T-lymphocyte-associated protein 4; LAG-3, lymphocyte-activation gene 3; TIGIT, T cell immunoreceptor with Ig and ITIM domains; FAK, focal adhesion kinase; SRC, proto-oncogene tyrosine-protein kinase; ULK1/2, unc-51-like kinase 1/2; CDK4/6, cyclin-dependent kinase 4/6; WEE-1, Wee1-like protein kinase; ATR, ataxia telangiectasia and Rad3-related protein. This figure was created using the tools provided by BioRender.com (accessed on 10 April 2024).
Ijms 25 04633 g002
Table 1. RAF and MEK inhibitors as monotherapies or in combination with other compounds under clinical evaluation in lung cancer. RAF, rapidly accelerated fibrosarcoma; BRAF, v-RAF murine sarcoma viral oncogene homolog B; MEK, mitogen-activated protein-kinase kinase; ERK, extracellular signal-regulated kinase; KRAS, Kirsten rat sarcoma viral oncogene homolog; mTOR, mechanistic target of rapamycin; CDK4/6, cyclin-dependent kinase 4/6; PD-1, programmed cell death protein 1; PD-L1, programmed death-ligand 1; VEGFR, vascular endothelial growth factor receptor; FGFR, fibroblast growth factor receptor; PDGFR, platelet-derived growth factor receptor; KIT, tyrosine-protein kinase; EGFR, epidermal growth factor receptor; RET, rearranged during transfection. Retrieved from clinicaltrials.gov (assessed on 27 February 2024).
Table 1. RAF and MEK inhibitors as monotherapies or in combination with other compounds under clinical evaluation in lung cancer. RAF, rapidly accelerated fibrosarcoma; BRAF, v-RAF murine sarcoma viral oncogene homolog B; MEK, mitogen-activated protein-kinase kinase; ERK, extracellular signal-regulated kinase; KRAS, Kirsten rat sarcoma viral oncogene homolog; mTOR, mechanistic target of rapamycin; CDK4/6, cyclin-dependent kinase 4/6; PD-1, programmed cell death protein 1; PD-L1, programmed death-ligand 1; VEGFR, vascular endothelial growth factor receptor; FGFR, fibroblast growth factor receptor; PDGFR, platelet-derived growth factor receptor; KIT, tyrosine-protein kinase; EGFR, epidermal growth factor receptor; RET, rearranged during transfection. Retrieved from clinicaltrials.gov (assessed on 27 February 2024).
Drug(s)Target(s)Second
Drug(s)
Second
Target(s)
Trial
Phase
Clinical Trial Identifier
BGB-3245RAF--INCT04249843
BDTX-4933RAF--INCT05786924
ExarafenibRAF--INCT04913285
FORE8394RAF--I/IINCT02428712
ABM-1310RAF+/− CobimetinibMEKINCT04190628
BelvarafenibRAFCobimetinib
Cetuximab *
MEK
EGFR
INCT03284502
BGB-3245RAFMirdametinibMEKI/IINCT05580770
DabrafenibΒRAFV600ETrametinibMEKIVNCT03340506
EncorafenibΒRAFV600EBinimetinibMEKIINCT03915951
EncorafenibΒRAFV600EBinimetinibMEKIINCT05195632
EncorafenibΒRAFV600EBinimetinibMEKIINCT04526782
EncorafenibΒRAFV600EBinimetinibMEKIINCT03839342
HLX208ΒRAFV600ETrametinibMEKINCT04965220
LifirafenibRAFMirdametinibMEKINCT03905148
NaporafenibRAFTrametinibMEKINCT05907304
NaporafenibRAFTrametinib
LTT462
Ribociclib
MEK
ERK
CDK4/6
INCT02974725
PF-07799933RAFBinimetinib
Cetuximab *
MEK
EGFR
INCT05355701
TinlorafenibRAFBinimetinibMEKINCT04543188
TovorafenibRAF+/− PimasertibMEKI/IINCT04985604
VemurafenibΒRAFV600ECobimetinibMEKII/IIINCT05768178
XP-102RAF+/− TrametinibMEKI/IINCT05275374
ABM-168MEK--INCT05831995
IMM-6-415MEK--I/IINCT06208124
IMM-1-104MEK(Chemotherapy)-I/IINCT05585320
AvutometinibMEK/RAFSotorasibKRASG12CI/IINCT05074810
AvutometinibMEK/RAF+/− EverolimusmTORINCT02407509
BinimetinibMEKPalbociclibCDK4/6I/IINCT03170206
BinimetinibMEKPembrolizumabPD-1INCT03991819
CobimetinibMEKAtezolizumabPD-L1IINCT03600701
MirdametinibMEKPalbociclibCDK4/6I/IINCT02022982
PF-07799544MEKTinlorafenib
PF-07799933
Encorafenib
RAFINCT05538130
SelumetinibMEKDocetaxel Microtubule
depolymerization
IIINCT01933932
SelumetinibMEKOsimertinibEGFRL858R/T790MIINCT03392246
TrametinibMEKAnlotinibVEGFR, FGFR, PDGFR, KITINCT04967079
TrametinibMEKEGF816EGFRINCT03516214
TrametinibMEKEverolimusmTORIINCT04803318
LenvatinibVEGFR, FGFR, PDGFR, KIT, RET
TrametinibMEKPembrolizumabPD-1INCT03299088
TrametinibMEKPembrolizumabPD-1I/IINCT03225664
TrametinibMEKDocetaxel Microtubule
depolymerization
IINCT02642042
* monoclonal antibody, chemotherapy medication.
Table 2. RAF and MEK inhibitors (highlighted) as secondary treatment or as sub-study treatment in multidrug testing trials under clinical evaluation in lung cancer. SOS1, son of sevenless homolog; MEK, mitogen-activated protein-kinase kinase; KRAS, Kirsten rat sarcoma viral oncogene homologue; PD-1, programmed cell death protein 1; TIGIT, T cell immunoreceptor with Ig and ITIM domains; BRAF, v-RAF murine sarcoma viral oncogene homolog B; VEGFR, vascular endothelial growth factor receptor; VEGF-A, vascular endothelial growth factor-A; PDGFRβ, platelet-derived growth factor receptor β; SHP2, Src homology 2 domain-containing phosphatase 2; ROS1, c-ros oncogene-1; MET, hepatocyte growth factor receptor; ALK, anaplastic lymphoma kinase; EGFR, epidermal growth factor receptor; CDK4/6, cyclin-dependent kinase 4/6; PD-L1, programmed death-ligand 1; CTLA-4, cytotoxic T-lymphocyte-associated protein 4; HER2, human epidermal growth factor receptor 2; RET, rearranged during transfection; WEE-1, Wee1-like protein kinase; AKT, protein kinase B; SRC, proto-oncogene tyrosine-protein kinase; BCR-ABL, breakpoint cluster region protein–tyrosine-protein kinase Abl1; FAK, focal adhesion kinase; FGFR1-4, fibroblast growth factor receptor 1-4; TRKA, tropomyosin receptor kinase A; PI3Kα,β,γ,δ, phosphatidylinositol 3-kinase-α,β,γ,δ; LAG-3, lymphocyte-activation gene 3; mTOR, mechanistic target of rapamycin; PI3KCA, phosphatidylinositol 3-kinase catalytic subunit alpha; SMO, smoothened; P-gp, permeability glycoprotein; ABCG2, ATP-binding cassette super-family G member; ATR, ataxia telangiectasia and Rad3-related protein. Retrieved from clinicaltrials.gov (assessed on 27 February 2024).
Table 2. RAF and MEK inhibitors (highlighted) as secondary treatment or as sub-study treatment in multidrug testing trials under clinical evaluation in lung cancer. SOS1, son of sevenless homolog; MEK, mitogen-activated protein-kinase kinase; KRAS, Kirsten rat sarcoma viral oncogene homologue; PD-1, programmed cell death protein 1; TIGIT, T cell immunoreceptor with Ig and ITIM domains; BRAF, v-RAF murine sarcoma viral oncogene homolog B; VEGFR, vascular endothelial growth factor receptor; VEGF-A, vascular endothelial growth factor-A; PDGFRβ, platelet-derived growth factor receptor β; SHP2, Src homology 2 domain-containing phosphatase 2; ROS1, c-ros oncogene-1; MET, hepatocyte growth factor receptor; ALK, anaplastic lymphoma kinase; EGFR, epidermal growth factor receptor; CDK4/6, cyclin-dependent kinase 4/6; PD-L1, programmed death-ligand 1; CTLA-4, cytotoxic T-lymphocyte-associated protein 4; HER2, human epidermal growth factor receptor 2; RET, rearranged during transfection; WEE-1, Wee1-like protein kinase; AKT, protein kinase B; SRC, proto-oncogene tyrosine-protein kinase; BCR-ABL, breakpoint cluster region protein–tyrosine-protein kinase Abl1; FAK, focal adhesion kinase; FGFR1-4, fibroblast growth factor receptor 1-4; TRKA, tropomyosin receptor kinase A; PI3Kα,β,γ,δ, phosphatidylinositol 3-kinase-α,β,γ,δ; LAG-3, lymphocyte-activation gene 3; mTOR, mechanistic target of rapamycin; PI3KCA, phosphatidylinositol 3-kinase catalytic subunit alpha; SMO, smoothened; P-gp, permeability glycoprotein; ABCG2, ATP-binding cassette super-family G member; ATR, ataxia telangiectasia and Rad3-related protein. Retrieved from clinicaltrials.gov (assessed on 27 February 2024).
Drug(s)Target(s)Second
Drug(s)
Second
Target(s)
Trial
Phase
Clinical Trial Identifier
BI-1701963SOS1+/− TrametinibMEKINCT04111458
DCC-3116ULK1/2Trametinib
Binimetinib
Sotorasib
MEK
MEK
KRASG12C
I/IINC04892017
SasanlimabPD-1Encorafenib/binimetinib
Axitinib
/SEA-TIGIT *
ΒRAFV600E
MEK
VEGFR1-3, PDGFRβ
TIGIT
I/IINCT04585815
PF-07284892SHP2Encorafenib
Binimetinib
Lorlatinib
Cetuximab *
ΒRAFV600E
MEK
ROS1, ALK
EGFR
INCT04800822
OpnurasibKRASG12CTrametinib
Ribociclib
Cetuximab *
MEK
CDK4/6
EGFR
I/IINCT05358249
Durvalumab *
Tremelimumab *
PD-L1
CTLA-4
SelumetinibMEKI/IINCT03581487
AlectinibALKCobimetinibMEKI/IINCT03202940
Caboplatin /pemetrexed /pembrolizumab *PD-1MirdametinibMEKI/IINCT05937906
Vemurafenib
Alectinib
Atezolizumab *
Trastuzumab emtansine #
BRAFV600E
ALK
PD-L1
HER2-microtubulin
--IINCT02314481
Atezolizumab *PD-L1Cobimetinib
/vemurafenib
MEK
BRAFV600E
VEGF-A
II/IIINCT03178552
Atezolizumab *PD-L1Bevacizumab *
/pemetrexed
/carboplatin
Atezolizumab *
Alectinib
Entrectinib
PD-L1
ALK
ROS1
DivarasibKRASG12CDocetaxel Microtubule
depolymerization
VemurafenibBRAFV600ECobimetinibMEKIINCT04302025
Alectinib
Entrectinib
Pralsetinib
Atezolizumab *
Divarasib
ALK
ROS1
RET
PD-L1
KRASG12C
--
DabrafenibBRAFV600ETrametinibMEKIINCT02465060
Binimetinib
Trametinib
Ulixertinib
Adavosertib
Afatinib
Capivasertib
Copanlisib
Crizotinib
Dasatinib
Defactinib
Erdafitinb
Fexagratinib
Ipatasertib
Larotrectinib
Osimertinib
Palbociclib
GSK2636771
Sapanisertib
Sunitinib
Taselisib
Vismodegib
MEK
MEK
ERK
WEE-1
EGFR, HER2
AKT
PI3Kα,β,γ,δ
ALK, MET, ROS1
SRC, BCR-ABL
FAK
FGFR1-4
FGFR1-4
AKT
TRKA, B and C
EGFRL858R/T790M
CDK4/6
PI3Kβ
mTOR
VEGFR2, PDGFRβ
PI3KCA, PI3Kα,β,γ,δ
SMO, P-gp, ABCG2
--
Pertuzumab *HER2Trastuzumab *HER2
Nivulomab *PD-1Relatlimab *LAG-3
SelumetinibMEKDocetaxel Microtubule
depolymerization
IINCT02664935
CeralasertibATRDurvalumab *PD-L1
SitravatinibAxl, MER, VEGFR1-3,
KIT, FLT3, DDR1-2, TRKA and B
Capivasertib
Fexagratinib
Vistusertib
Palbociclib
Crizotinib
Osimertinib
AKT
FGFR1-4
mTOR
CDK4/6
ALK, MET, ROS1
EGFRL858R/T790M
* monoclonal antibody, chemotherapy medication, # antibody–drug conjugate.
Table 3. Key clinical trial outcomes for BRAFV600E-mutant NSCLC patients upon treatment with RAF and/or MEK inhibitors and their most frequent adverse events. ORR, objective response rate; TRAEs, treatment-related adverse events.
Table 3. Key clinical trial outcomes for BRAFV600E-mutant NSCLC patients upon treatment with RAF and/or MEK inhibitors and their most frequent adverse events. ORR, objective response rate; TRAEs, treatment-related adverse events.
Drug(s)Target(s)Number
of Patients
ORRTRAEsReference
Encorafenib
+ binimetinib
BRAFV600E
MEK
9875%Nausea, diarrhea,
fatigue
[91]
Dabrafenib
+ trametinib
BRAFV600E
MEK
9368.4%Pyrexia, nausea,
vomiting
[87]
Dabrafenib
+ trametinib
BRAFV600E
MEK
3664%Pyrexia, nausea,
fatigue,
peripheral oedema
[86]
DabrafenibBRAFV600E8433%Pyrexia,
hyperkeratosis,
decreased appetite
[47]
VemurafenibBRAFV600E10144.9%Nausea,
hyperkeratosis,
decreased appetite
[66]
Cobimetinib
+ atezolizumab *
MEK
PD-L1
2818%Diarrhea, rash,
fatigue
[94]
* monoclonal antibody.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Adamopoulos, C.; Papavassiliou, K.A.; Poulikakos, P.I.; Papavassiliou, A.G. RAF and MEK Inhibitors in Non-Small Cell Lung Cancer. Int. J. Mol. Sci. 2024, 25, 4633. https://doi.org/10.3390/ijms25094633

AMA Style

Adamopoulos C, Papavassiliou KA, Poulikakos PI, Papavassiliou AG. RAF and MEK Inhibitors in Non-Small Cell Lung Cancer. International Journal of Molecular Sciences. 2024; 25(9):4633. https://doi.org/10.3390/ijms25094633

Chicago/Turabian Style

Adamopoulos, Christos, Kostas A. Papavassiliou, Poulikos I. Poulikakos, and Athanasios G. Papavassiliou. 2024. "RAF and MEK Inhibitors in Non-Small Cell Lung Cancer" International Journal of Molecular Sciences 25, no. 9: 4633. https://doi.org/10.3390/ijms25094633

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop