Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2021

Open Access 01.12.2021 | Review

MEK inhibitors for the treatment of non-small cell lung cancer

verfasst von: Jing Han, Yang Liu, Sen Yang, Xuan Wu, Hongle Li, Qiming Wang

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2021

Abstract

BRAF and KRAS are two key oncogenes in the RAS/RAF/MEK/MAPK signaling pathway. Concomitant mutations in both KRAS and BRAF genes have been identified in non-small cell lung cancer (NSCLC). They lead to the proliferation, differentiation, and apoptosis of tumor cells by activating the RAS/RAF/MEK/ERK signaling pathway. To date, agents that target RAS/RAF/MEK/ERK signaling pathway have been investigated in NSCLC patients harboring BRAF mutations. BRAF and MEK inhibitors have gained approval for the treatment of patients with NSCLC. According to the reported findings, the combination of MEK inhibitors with chemotherapy, immune checkpoint inhibitors, epidermal growth factor receptor-tyrosine kinase inhibitors or BRAF inhibitors is highly significant for improving clinical efficacy and causing delay in the occurrence of drug resistance. This review summarized the existing experimental results and presented ongoing clinical studies as well. However, further researches need to be conducted to indicate how we can combine other drugs with MEK inhibitors to significantly increase therapeutic effects on patients with lung cancer.
Hinweise
Jing Han and Yang Liu contributed equally to this work

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
NSCLC
Non-small cell lung cancer
RTK
Receptor tyrosine kinase
GRB
Growth factor receptor bound protein
SOS
Son of Sevenless homolog
GDP
Guanosine diphosphate
GTP
Guanosine triphosphate
RAS
Rat sarcoma viral oncogene
RAF
V-raf murine sarcoma viral oncogene
MEK
Mitogen-activated protein kinase kinase
ERK
Extracellular signal-regulated kinase
PI3K
Phosphatidylinositol 3-kinase
AKT
Protein kinase B
mTOR
Mammalian target of rapamycin
NF-kB
Nuclear factor-kB
NA
Non-available
ORR
Objective response rate
OS
Overall survival
PFS
Progression-free survival
HR
Hazard ratio
MEKi
MEK inhibitors
BRAFi
BRAF inhibitors
CT
Chemotherapy
ICI
Immune checkpoint inhibitors
EGFR-TKI
Epidermal growth factor receptor tyrosine kinase inhibitors
DLTs
Dose-limiting toxicities
NCCN
National Comprehensive Cancer Network
FDA
The United States Food and Drug Administration
EMA
European Medicines Agency

Introduction

Lung cancer is the most common cause of cancer-related death worldwide, with over 1.8 million lung cancer deaths annually [1]. Over the past decades, the treatment of non-small cell lung cancer (NSCLC) has changed dramatically with the development of molecular profiling, targeted therapeutic agents, and precision medicine, while the overall prognosis of lung cancer is still poor with a 5-year overall survival (OS) rate of 18% across all stages [2]. NSCLC accounts for about 80–85% of lung cancer cases and almost 70% of NSCLC patients presenting with locally advanced or metastatic disease at initial diagnosis [3]. NSCLC comprises several histologic subtypes, such as squamous cell carcinoma, adenocarcinoma, large cell or undifferentiated carcinoma. Non-squamous carcinoma (70–75%) and squamous cell carcinoma (25–30%) are two major subtypes [4]. In NSCLC somatic mutations in epidermal growth factor receptor (EGFR) and rearrangements in anaplastic lymphoma kinase gene (ALK) and ROS proto-oncogene1 (ROS1) have been validated as strong predictive biomarkers and attractive drug targets. However, the mitogen-activated protein kinase (MAPK) pathway, comprising the kinases RAS, RAF, MEK, and ERK, is also implicated in the tumorigenesis of NSCLC. Thus, MEK inhibitors’ monotherapy or combination with other targeted drugs harboring MAPK pathway become a promising strategy for NSCLC patients with B-Raf proto-oncogene (BRAF) or Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations. Currently, the prevalence of BRAF mutations is 3–5% in NSCLC patients, of which BRAF V600E mutations constitute approximately 50% [5]. To date, BRAF plus MEK inhibitors have shown a remarkable survival and response rate in advanced and unresectable melanoma patients, compared with single-agent BRAF inhibition [6, 7]. Moreover, concomitant inhibition of both BRAF and MEK has been validated to overcome acquired resistance to BRAF inhibitors alone [8, 9]. Besides, the prevalence of KRAS mutations is ~ 25% and ~ 15% in Western and Asian populations with lung adenocarcinoma, respectively [10]. Although the unprecedented challenge of effective KRAS targeting is evidenced by disappointing results to date, MEK inhibitors plus other targeted agents are actively exploring the potential effect in some clinical trials right now.
The present study aimed to review researches concentrated on the effects of MEK inhibitors on NSCLC patients to facilitate the clinical management of such patients.

Structures and functions of MEK proteins

MEK proteins are mitogen-activated protein kinase kinase, a dual specificity Tyr/Thr protein kinase that selectively phosphorylates serine/threonine and tyrosine residues in the activation loop of ERK1 and ERK2. MEK proteins are coded by 7 different genes, among which MEK1 and MEK2 are of significance. MEK1 gene exists in human chromosome 15q22.31, and MEK2 gene exists in chromosome 9q13.3 [11]. The MEK1/2 proteins have three crucial domains (Fig. 1): a core protein kinase domain, an N-terminal domain (approximately 80 amino acids), and a shorter C-terminal region (within 30 amino acids) [11, 12]. The protein kinase domain contains the ATP site and catalytic segment; besides, a pocket structure near the ATP-binding site is an ideal target for small target agents that can change the molecule to an inactive state. The N-terminal region plays a regulatory role in signal transduction, including the D-domain (docking site) binding to the ERK substrate. Additionally, mitogen-activated protein kinase (MAPK) is localized to the cytoplasm through its specific association with the N-terminal 1–32 residues of MAPKK in unstimulated cells [13]. The C-terminal region contains the domain for versatile docking (DVD), a critical binding site for the upstream apparatus of the MAPK signaling pathway [14].

Molecular pathways and MEK inhibitors

MEK is the downstream of RAS/RAF/MEK/ERK signaling pathway, highly regulating and playing an important role in cell proliferation, differentiation, apoptosis, and stress responses [15]. It transmits mitogenic signals from outside the cell to the nucleus through multistage phosphorylation [16]. In tumor cells, certain growth factors are combined with transmembrane receptors on the cell surface, leading to the increase in RAS guanosine triphosphate-binding protein in the cell [17]. Once RAS is activated, the plasma membrane of the cell secretes and activates the downstream molecule RAF kinase, stimulates a series of protein kinases, and forms the RAS/RAF/MEK/ERK signaling pathway [18] (Fig. 2).
To date, four MEK inhibitors have been approved by the United States Food and Drug Administration (FDA), including trametinib, binimetinib, selumetinib, and cobimetinib [1922]. They are oral, allosteric, selective, ATP-non-competitive MEK1/2 inhibitors that are not easy to produce cross-inhibition to other targets [2327]. Notably, trametinib is the only MEK inhibitor approved for the treatment of NSCLC patients with BRAF V600E mutation in combination with dabrafenib till now (Table 1).
Table 1
Approval and status of MEK inhibitors in active clinical development
Drug
Developer or owner
Target
In vitro IC50 for MEK (nM)
Tumor
Approval/development status
Trametinib (GSK1120212, JTP-74057)
NOVARTIS
MEK1/2
0.7 (MEK1), 0.9 (MEK2) [24]
Melanoma, NSCLC, thyroid cancer
Approved by US FDA (05/2013)
Binimetinib (MEK162, ARRY-438162)
ARRAY BIOPHARMA INC
MEK1/2
12 [28]
Melanoma
Approved by US FDA (06/2018)
Selumetinib (AZD6244, ARRY-142886)
ASTRAZENECA
MEK1/2
14 [23]
Neurofibroma
Approved by US FDA (4/2020)
Cobimetinib (GDC-0973, XL518)
GENENTECH INC
MEK1/2
5 [29]
Melanoma
Approved by US FDA (11/2015)
Pimasertib (AS703026, MSC1936369B)
Merck KGaA
MEK1/2
30 [30]
Melanoma, ovarian cancer, pancreatic adenocarcinoma, solid tumor
I/II
Mirdametinib (PD-0325901)
Spring Works Therapeutics
MEK1/2
0.1–1000 [31]
Neurofibroma, solid tumor
I/II
Refametinib (BAY 86–9766, RDEA119)
Bayer AG
MEK1/2
19 (MEK1), 47 (MEK2) [32]
Biliary tract cancer, hepatocellular cancer, solid tumor
I/II
E6201
Eisai Co Ltd./Strategia Theraputics
MEK1/FLT3
NA
Melanoma with brain metastases, solid tumor
I
GDC-0623 (RG 7421)
Genentech, Inc.
MEK1
0.13 [33, 34]
Solid tumor
I
CH5126766(RO5126766)
Chugai Pharmaceutical Co., Roche
MEK/BRAF/CRAF
160/190/56 [35]
KRAS-mutant NSCLC, solid tumor
I
HL-085
Shanghai Kechow Pharma, Inc.
MEK1/2
1.9–10 [27]
Melanoma, NSCLC, solid tumor
I/II
SHR7390
HENGRUI MEDICINE
MEK1/2
NA
Breast neoplasm, solid tumor
I/II
TQ-B3234
CHIATAI TIANQING
MEK1/2
NA
Solid tumor
I
CS-3006
CSTONE PHARMACEUTICALS
MEK1/2
NA
Solid tumor
I
FCN-159
FOSUN PHARMA
MEK1/2
NA
NRAS-aberrant (Ia) and NRAS-mutant (Ib) melanoma
I

Evidence for MEK monotherapy for NSCLC patients

Several trials have explored the function of single-agent MEK inhibition in early clinical development. An initial phase II study evaluated the efficacy and safety of AZD6244 versus pemetrexed as second- or third-line treatment in patients with advanced NSCLC. In this trial, 84 patients were enrolled, and 5% and 4.5% of patients achieved an objective response in AZD6244 group and pemetrexed group, respectively. However, there was no significant difference in median progression-free survival (PFS) between the two groups (90 days vs 67 days, HR:1.08, P = 0.79). The incidence of treatment-related serious adverse events appeared more commonly in the pemetrexed group (6.8% vs 2.5%) than in the AZD6244 group. Most frequently, toxicities were primarily dermatitis acneiform (43%), diarrhea (30%), nausea (18%), and vomiting (18%) with AZD6244 [36]. Another single-arm phase II study was conducted to test PD-0325901 in two administration schedules. This study enrolled 34 patients. Thirteen patients were administered intermittently (3 weeks on/1 week off), while 21 patients were administered adjusting schedule (5 days on/2 days off for 3 weeks, followed by 1 week off). No objective responses were observed in two schedules, while 7 patients had stable disease. Median PFS was 1.8 months (95% CI 1.5–1.9), and overall survival was 7.8 months (95% CI 4.5–13.9). The most common treatment-related toxicities (incidence in schedule A/incidence in schedule B) were diarrhea (54%/76%), fatigue (31%/48%), rash (46%/33%), vomiting (38%/33%), and nausea (38%/29%) [37]. Another phase II study evaluated the safety and efficacy of trametinib versus docetaxel for patients with KRAS-mutant NSCLC patients. In this trial, 129 were enrolled. However, there was no significant difference in median PFS in trametinib and docetaxel arm (12 weeks vs 11 weeks, HR:1.14, P = 0.5197) and in median OS (8 months vs not reached, HR:0.97, P = 0.934). Partial responses (PRs) for these two groups were 12% and 12% (P = 1.0000). The most frequent grade 3 or higher toxicities were primarily hypertension (9%), rash (9%), diarrhea (5%), sepsis (5%), and asthenia (5%) vs. neutropenia (35%) in trametinib and docetaxel arms, respectively. One treatment-related death occurred with trametinib and none with docetaxel [38]. An initial phase II basket trial evaluated the efficacy of selumetinib in NSCLC patients with molecular profiling. In this trial, 110 patients presented RAS/RAF mutations with KRAS mutations (24.9%), BRAF mutations (2%), HRAS and NRAS mutations (0.7%), and 10 patients were enrolled onto the selumetinib arm. However, 9 patients failed to achieve selumetinib monotherapy primary end point, with only one partial response (ORR 11%, 95% CI 0–48%), a median PFS time of 2.3 months, and median OS time of 6.5 months [39]. The results of these phase II studies indicated that MEK inhibitors’ monotherapy seemed to have poor clinical outcomes and more toxicities for NSCLC patients compared with chemotherapy alone.

Evidences for combination of BRAF and MEK inhibitors for NSCLC patients

The combination of BRAF and MEK inhibitors has been proved to be clinically effective for NSCLC patients to date. An initial phase II trial evaluated the combination of dabrafenib and trametinib in previously treated BRAF(V600E)-mutant NSCLC patients. Fifty-seven patients were enrolled in this study. The overall response was 63.2% (95% CI 49.3–75.6%), the median PFS was 9.7 months (95% CI 6.9–19.6), and median duration of response (DOR) was 9.0 months (95% CI 6.9–18.3). Common grade 3/4 AEs were neutropenia (9%), hyponatremia (7%), and anemia (5%) [40]. Besides, the same research team developed another phase II study to assess the efficacy and safety of dabrafenib plus trametinib treatment in previously untreated patients with BRAF(V600E)-mutant metastatic NSCLC. In this study, 36 patients were enrolled and treated with first-line dabrafenib plus trametinib. The ORR was 64% (95% CI 46–79%), median DOR was 10.4 months (95%CI 8.3–17.9), and PFS was 10.9 months (95% CI 7.0–16.6). Grade 3 or 4 AEs were pyrexia (11%), alanine aminotransferase increase (11%), hypertension (11%), and vomiting (8%) [41]. The NCI-MATCH Trial Subprotocol H evaluated the combination of dabrafenib and trametinib in solid tumor patients, 5 lung adenocarcinoma patients included. One patient was progression-free at 32.5 months, and 1 patient who was considered unevaluable, with an 81% reduction in the sum of measured lesions, had a PFS of 12.7 months. Three patients had SD for 15.6, 6.6, and 3.6 months which is sought to investigate the selective BRAF inhibitor [42]. The clinical data showed the efficacy of combination of MEK and BRAF inhibitors with untreated or treated BRAF V600E-mutant metastatic NSCLC, indicating that physicians can flexibly treat patients with this targeted therapy combination in either the first-line or following chemotherapy and provide strategies to accommodate the individual patient needs.

Evidence for combination of chemotherapy and MEK inhibitors for NSCLC patients

Chemotherapy is no longer the most efficacious treatment, and targeted agents have been rationally designed to inhibit particular mutations, leading to a more streamlined clinical trial process. Ten years ago, numerous clinical trials have concentrated on exploration of the combination of chemotherapy plus MEK inhibitors for NSCLC patients (Table 2). In the early stage, a phase II study evaluated selumetinib plus docetaxel versus docetaxel plus placebo for patients with KRAS-mutant advanced NSCLC. Forty-four and 43 patients were enrolled in selumetinib and placebo groups, respectively. The median OS was 5.2, 9.4 months (HR: 0.80, P = 0.21) in selumetinib and placebo group, respectively. However, the median PFS in the selumetinib group was significant longer than the placebo group (5.3 months vs. 2.1 months, HR: 0.58, P = 0.014). Similarly, the ORR was 37% and none (P < 0.0001) in selumetinib and placebo groups, respectively. Grade 3 or higher AEs occurred in 82% patients in selumetinib group and 67% patients in the placebo group (Table 3) [43]. Another phase II study of selumetinib in combination with chemotherapy was conducted in patients with advanced or metastatic non-squamous NSCLC. A total of 63 enrolled patients were randomly assigned 1:1:1 to intermittent selumetinib + chemotherapy (arm A) or continuous selumetinib + chemotherapy (arm B) or chemotherapy alone (arm C). The ORR was 35%, 62%, and 24% in arm A/B/C, respectively. Similarly, the PFS was 7.5, 6.7, 4.0 respectively. Skin and gastrointestinal adverse events were more common with the addition of selumetinib (Table 3) [44]. A phase II trial evaluating the combination of selumetinib plus docetaxel in KRAS-mutant advanced NSCLC patients also demonstrated modest improved efficacy. The retrospective analysis indicated that OS for the selumetinib + docetaxel arm vs. placebo + docetaxel arm in KRAS mutation group (MG1) and MG2 was 9.6 vs 4.4 months and 8.6 vs 7.1 months, respectively. Similarly, PFS for selumetinib and placebo groups in KRAS MG1 and MG2 was 5.7 vs 1.4 months and 4.9 vs 2.6 months, respectively. The ORR showed a numerically higher rate in MG1 compared with MG2 (46% vs 26%, respectively). Thus, for patients receiving selumetinib‏ + docetaxel and harboring KRAS G12C or G12V mutations, there were trends toward greater improvement in OS, PFS, and ORR compared with other KRAS mutations [45]. A phase 1/1b study evaluated the efficacy and safety of trametinib plus docetaxel or pemetrexed in advanced NSCLC. In this trial, 95 patients were enrolled. In trametinib plus docetaxel group, the ORR was 18% versus 24% in KRAS-WT and KRAS-mutant, respectively. In trametinib plus pemetrexed group, the ORR was 17% versus 11% in KRAS-WT and KRAS-mutant, respectively. Most common AEs were diarrhea, nausea, and fatigue (Table 3) [46]. SELECT-1 was designed to assess the efficacy and safety of selumetinib plus docetaxel in patients with KRAS-mutant locally advanced or metastatic NSCLC. In total, 510 patients were enrolled and randomized. PFS was 3.9, 1.1 months in selumetinib and placebo groups, respectively (HR:0.93, P = 0.44). OS was 8.7, 0.9 months (HR:1.05, P = 0.64), respectively. ORR was 20.1% and 13.7% in selumetinib and placebo groups, respectively. Grade 3 or higher AEs were more frequent with selumetinib group than placebo (67% vs 45%) (Table 3) [47]. However, Jacob Kaufman et al. from Duke University questioned whether other mutations are related to the response to MEK inhibition, such as the concurrent loss of tumor-suppressor genes in LKB1, which may also affect the results of the trial [48]. The SELECT-2 trial assessed the efficacy of selumetinib plus docetaxel as a second-line treatment for patients with advanced metastatic NSCLC. A total of 212 patients were randomized. There were no statistically significant improvements in PFS or OS for overall or KRAS-WT in either selumetinib or placebo group. PFS for selumetinib + docetaxel 60 mg/m2, selumetinib + docetaxel 75 mg/m2 compared with placebo + docetaxel 75 mg/m2 was 3.0, 4.2, and 4.3 months. The most commonly reported grade 3 or higher AE was neutropenia (Table 3) [49]. SELECT-3 trial was designed a phase I study to assess the efficacy of selumetinib in combination with platinum-doublet chemotherapy for NSCLC patients in first-line setting. Fifty-five patients were enrolled. Most frequent adverse events (AEs) were fatigue, nausea, diarrhea, and vomiting (Table 3) [50]. Another phase I study evaluated the safety and tolerability of selumetinib as a monotherapy, or in combination with docetaxel as a second-line therapy for Japanese patients with advanced NSCLC. Thirty-three patients were enrolled and 25 assigned to treatment. Grade 3 dose-limiting toxicities were febrile neutropenia and pneumonitis (Table 3) [51]. Current clinical data showed that MEK inhibitor combined with chemotherapy can improve the outcomes while some not. One possibility is that clinical benefit may occur in a specific subset of tumors that exhibits a favorable genetic of signaling environment. So effective drug candidates of MEK inhibitors and proper special patients should be detected for this combination therapy.
Table 2
Completed clinical trials of chemotherapy + MEK inhibitors in NSCLC
Study
Study design
Intervention
Comparation
Patient population
Patients (n)
Median OS (months)
Median PFS (months)
ORR (%)
Jänne et al. [43]
Phase2 (NCT00890825)
Selumetinib + docetaxel
Placebo + docetaxel
KRAS-mutant advanced NSCLC
87 (44 vs 43)
9.4 vs 5.2 (HR:0.8, 80% CI = 0.56–1.14, P = 0.21)
5.3 vs 2.1 (HR:0.58, 80%CI = 0.42–0.79, P = 0.014)
37% vs 0
Gandara et al. [46]
Phase1 (NCT01192165)
Trametinib + docetaxel
Trametinib + pemetrexed
NSCLC
95 (49 vs 46)
NA
KRAS wild-type:4.2 vs 5.8
KRAS-mutant type: 3.4 vs 4
KRAS wild-type:18% vs 11%
KRAS-mutant type: 24% vs 17%
Jänne et al. [47]
Phase1 (NCT01933932)
Selumetinib + docetaxel
Placebo + docetaxel
KRAS-mutant NSCLC
510 (251 VS 254)
8.7 VS 7.9 (HR:1.05, 95% = 0.85–1.30, P = 0.64)
3.9 VS 2.8 (HR:0.93, 95%CI = 0.77–1.12, P = 0.44)
20.1% vs 13.7% (OR:1.61, 95%CI = 1–2.62, P = 0.05)
Soria et al. [49]
Phase2 (NCT01750281.)
Selumetinib + docetaxel
Placebo + docetaxel
NSCLC
212
5.7 vs 7.7 vs 11.5
3 vs 4.2 vs 4.3 (HR = 1.12,0.92)
33% vs 14% (OR:3.26, 95%CI = 1.47–7.95)
Greystoke et al. [50]
Phase1 (NCT01809210)
Selumetinib + gemcitabine/cisplatin or carboplatin
Selumetinib + pemetrexed/cisplatin or carboplatin
NSCLC
55
NA
NA
36% vs 33% vs 19% vs 13%
Seto et al. [51]
Phase1 (NCT01605916)
Selumetinib + docetaxel
Selumetinib
Solid tumor of NSCLC
25
NA
NA
NA
Melosky et al. [44]
phase2
Selumetinib + pemetrexed + cisplatin
No selumetinib
Non-squamous NSCLC
62
10 vs 10.1 vs 15.3 (HR = 1.56,1.72)(P = 0.31,0.2)
7.2 vs 6.9 vs 4 (HR = 0.82,0.77)(P = 0.56,0.44)
35% vs 62% vs 24%
NA, non-available; NSCLC, non-small cell lung cancer; ORR, objective response rate; OS, overall survival, PFS, progression-free survival; HR, hazard ratio
Table 3
Main grade 3 or higher adverse events
Study
Main grade 3 or higher adverse events (over 10%)
Jänne et al. [43]
Neutropenia (67%), febrile neutropenia (18%), dyspnea (2%), and asthenia (9%)
Gandara et al. [46]
Trametinib + docetaxel: anemia (16%), asthenia (4%), diarrhea (10%), dyspnea (4%), fatigue (10%), hypoalbuminemia (4%), mucosal (4%), neutropenia (22%), and stomatitis (4%). Trametinib + pemetrexed: anemia (13%), AST increased (4%), asthenia (9%), decreased appetite (4%), diarrhea (4%), dyspnea (7%), hyponatremia (15%), nausea (7%), and neutropenia (20%)
Jänne et al. [47]
Diarrhea (6%), rash (3%), nausea (1%), fatigue (2%), stomatitis (3%), edema peripheral (1%), vomiting (2%), asthenia (5%), decreased appetite (1%), dermatitis acneiform (2%), neutropenia (6%), anemia (1%) and dyspnea (2%)
Greystoke et al. [50]
Neutropenia (26%), anemia (22%), and thrombocytopenia (20%)
Seto et al. [51]
Selumetinib monotherapy: blood and lymphatic system disorders (6%), neutropenia (6%), investigations (18%), AST increase (6%), GGT increase (6%), WBC count decrease (6%), infections and infestations (6%), pneumonia (6%), gastrointestinal disorders (12%), diarrhea (6%), vomiting (6%), respiratory, thoracic and mediastinal disorders (12%), interstitial lung disease (6%), metabolism and nutrition disorders (6%) and hypoalbuminemia (6%)

Evidence for combination of immune checkpoint inhibitors and MEK inhibitors for NSCLC patients

Immune checkpoint inhibitors (ICIs) have opened up a new era for lung cancer treatment in recent years. However, even when patients with 50% or higher positivity for PD-L1 expression are selected, overall response rates still do not exceed 31% [52, 53]. Thus, different combination treatments have been proposed. Preclinical data suggested an improved T cell activation and increased CTLA-4 expression for selumetinib and trametinib. Besides, pulsatile MEKi treatment combined with CTLA-4 blockade prolonged survival in mice-bearing tumors with mutant KRAS [54, 55]. An initial phase Ib study was conducted to investigate the safety and efficacy of cobimetinib plus atezolizumab for patients with solid tumors (n = 152), 28 NSCLC patients included. The median OS time was 13.2 months, and ORR was 18% with NSCLC. 12-month PFS and OS rates were 29% and 57% for NSCLC patients, respectively. The most common AEs were diarrhea (67%), skin rash (48%), and fatigue (40%) [56]. Another phase I/II trial evaluated immunotherapy with durvalumab and tremelimumab with continuous or intermittent administration of selumetinib in NSCLC patients. The trial is actively screening and enrolling patients, and the estimated study completion is scheduled for April 2021 [57]. Currently, the clinical data about ICI-combined MEK inhibitors are still not efficient enough to validate the most proper way to treat NSCLC. More clinical outcomes are worthy being awaited furthermore.

Evidence for combination of EGFR tyrosine kinase inhibitors (TKIs) and MEK inhibitors for NSCLC patients

To our knowledge, acquired resistance has become a major clinical problem for advanced NSCLC patients with the increasing administration of EGFR-TKIs. The combination strategy of MEK inhibitors plus EGFR-TKIs has been proposed in certain clinical trials. Preclinical data suggested the stronger inhibitory effect of the cell proliferation of EGFR-TKIs-resistant cells for MEK inhibitors plus EGFR-TKIs [58]. A phase II study was concentrated on administration of selumetinib with and without erlotinib for KRAS-mutant and KRAS wild-type (WT) advanced NSCLC patients. Forty-one KARS-mutant and 38 KRAS-WT patients were enrolled. In KRAS-WT cohort, the median PFS was 2.1 and 2.4 months for erlotinib + selumetinib and erlotinib, respectively. Similarly, OS was 12.9 and 6.3 months, respectively. In KRAS-mutant cohort, the median PFS was 2.3 and 4.0 months for erlotinib + selumetinib and selumetinib, respectively. Similarly, OS was 21.8 and 10.5 months, respectively. In terms of safety, grade 3 and 4 toxicities were also increased in combination therapy, with diarrhea, dehydration, and fatigue all occurring in > 20% of patients [59]. TATTON was initially designed as a phase Ib trial to assess the safety and tolerability of osimertinib in combination with selumetinib, savolitinib, or durvalumab for EGFR-mutant lung cancer patients. Seventy-seven patients were enrolled in this study. The ORR was 42%, 44%, and 43% in selumetinib + osimertinib, savolitinib + osimertinib, and durvalumab + osimertinib arms, respectively. The most common AEs in selumetinib plus osimertinib group were diarrhea (75%), skin rash (58%), nausea (47%) [60]. Another phase I study evaluated the efficacy of afatinib plus selumetinib in patients with KRAS-mutant-positive solid tumors, 6 NSCLC patients included. Dose-limiting toxicities (DLTs) consisted of grade 3 diarrhea, decreased appetite, nausea/vomiting, dehydration, and mucositis. Stable disease for 221 days in a NSCLC patient was the best response [61]. In ESMO 2019 Congress, a phase I study evaluated the combination of lapatinib and trametinib for patients with KRAS-mutant solid tumors, 15 NSCLC patients included. One patient was confirmed partial response. Grade 3 AEs were diarrhea, rash, and nausea [62]. The clinical data showed that a number of trials were focused on detecting the strong rationale supporting combination therapy with MEK inhibitors for overcoming or delaying drug resistance in EGFR-mutant NSCLC. However, there are no EGFR-based combination therapies with global adoption, and therapies for patients with acquired resistance to EGFR-TKIs remain to be detected.

Mechanisms of resistance to MEK inhibitors

RAS/RAF/MEK/ERK signaling pathway-associated inhibitors have proven to be effective in treatment of various types of cancer, but have presented drug resistance in clinical application and MEK inhibitors as well. The resistance mechanisms to MEK inhibitors have not been detected clearly to date. However, studies concentrated on metastatic melanoma and other tumors showed some underlying mechanisms expected to be overlapped. A large number of MEK-acquired drug resistance mutations have been detected, such as the acquired concurrent MEK2-Q60P mutation and BRAF V600E amplification, which conferred resistance to MEK and BRAF inhibitors [63]. MEK1P124 and MEK1Q56P mutations were evaluated to be the mechanism of cross-resistance to PLX4720 (a selective BRAF inhibitor) and selumetinib [64]. Moreover, RAS can simultaneously induce ERK/MAPK and PI3K/AKT signaling pathways to induce drug resistance to MEK inhibitors. In preclinical studies [6569], the combination of inhibitors, such as mTOR, PI3K, AKT/Raf, and dual inhibitors of RTK/MAPK and PI3K/AKT signaling pathways was proved to be effective to overcome drug resistance of MEK inhibitors. Besides, tumor microenvironment (TME) has been detected to play a pivotal role in promotion of the targeted therapy resistance as well [70].

Other combined therapies and ongoing studies

As acquired resistance becomes a frequent problem for all the target agents, a number of clinical trials have been designed to evaluate the efficacy and safety of combination of two different types of targets plus MEK inhibitors, according to the probable resistance mechanisms in the former part. A preclinical experiment revealed that selumetinib combined with BEZ235 (PI3K/mTOR inhibitor) markedly enhanced their antitumor effects and inhibited the tumor growth of NCI-H1993 in gefitinib-resistant NSCLC xenograft models [71]. Other ongoing clinical trials on administration of MEK inhibitors for NSCLC patients have been summarized (Table 4). To date, a variety of MEK1/2 inhibitors have been applied for different types of cancer, including NSCLC at various stages of clinical testing. The publication of the final results of these studies is still awaited.
Table 4
Ongoing MEK inhibitors’ clinical trials in NSCLC
Trial NCT number
Intervention
Cancer type
Phase
Status
03170206
Binimetinib + Palbociclib
KRAS-mutant NSCLC
I/II
Recruiting
01859026
Erlotinib + Binimetinib
KRAS- or EGFR-mutant NSCLC
I/IB
Active, not recruiting
02185690
Carboplatin + Pemetrexed + Binimetinib
NSCLC
I
Active, not recruiting
02964689
Cisplatin + Pemetrexed + Binimetinib
KRAS-mutant NSCLC
I
Active, not recruiting
03581487
Durvalumab + Selumetinib + Tremelimumab
NSCLC
I/II
Recruiting
03991819
Binimetinib + Pembrolizumab
NSCLC
I
Active, not recruiting
01586624
Selumetinib + Vandetanib
NSCLC
I
Active, not recruiting
04526782
Encorafenib + Binimetinib + Docetaxel
BRAF V600E-mutant NSCLC
II
Not yet recruiting
01336634
Dabrafenib + Trametinib
BRAF V600E-mutant NSCLC
II
Active, not recruiting
04005144
Brigatinib + Binimetinib
ALK or ROS1 rearranged NSCLC
I
Recruiting
03087448
Ceritinib + Trametinib
ALK-positive NSCLC
I/II
Recruiting
01933932
Selumetinib + Docetaxel
KRAS-mutant NSCLC
III
Active, not recruiting
03600701
Atezolizumab + Cobimetinib
NSCLC
II
Recruiting
03202940
Alectinib + Cobimetinib
ALK rearranged NSCLC
IB/II
Recruiting
02642042
Trametinib + Docetaxel
KRAS-mutant stage IV NSCLC
II
Active, not recruiting
03299088
Pembrolizumab + Trametinib
KRAS-mutant
NSCLC
I
Recruiting
03516214
EGF816 + Trametinib
EGFR-mutant NSCLC
I
Recruiting
03225664
Trametinib + Pembrolizumab
NSCLC
I/II
Recruiting
01750281
Selumetinib + Docetaxel
NSCLC
II
Active, not recruiting
02664935
National Lung Matrix Trial: AZD4547/Vistusertib/Palbociclib/ Crizotinib/ Selumetinib/Docetaxel/AZD5363/ Osimertinib/ Durvalumab/ Sitravatinib/AZD6738
NSCLC
II
Recruiting
03990077
HL-085 + Docetaxel
KRAS-mutant
NSCLC
I
Not yet recruiting
01912625
Trametinib + Carboplatin + Paclitaxel + Radiation Therapy
NSCLC
I
Active, not recruiting
Data source: www.​clinicaltrials.​gov, cutoff data: October 24, 2020

Conclusions/expectations

The functions of EGFR-TKIs, checkpoint inhibitors, and traditional chemotherapy have been widely studied in NSCLC patients, while the role of MEK inhibitors in the treatment of lung cancer has not been clearly described. A number of clinical trials explored the clinical application of MEK inhibitors, and combination therapy has demonstrated promising outcomes. The brief summarization of MEK inhibitors in the selected clinical trials with NSCLC can be found in Table 5.
Table 5
MEK inhibitors in clinical trials
Study
Phase
MEK inhibitors
Drug therapy
Hainsworth et al. [36]
Phase II
Selumetinib
MEKi
Haura et al. [37]
Phase II
Mirdametinib
MEKi
Blumenschein et al. [38]
Phase II
Trametinib
MEKi
Lopez-Chavez et al. [39]
Phase II
Selumetinib
MEKi
Planchard et al. [40]
Phase II
Trametinib
MEKi + BRAFi
Planchard et al. [41]
Phase II
Trametinib
MEKi + BRAFi
Salama et al. [42]
Phase II
Trametinib
MEKi + BRAFi
Jänne et al. [43]
Phase II
Selumetinib
MEKi + CT
Gandara et al. [46]
Phase I/ Ib
Trametinib
MEKi + CT
Jänne et al. [47]
Phase III
Selumetinib
MEKi + CT
Soria et al. [49]
Phase II
Selumetinib
MEKi + CT
Greystoke et al. [50]
Phase I
Selumetinib
MEKi + CT
Seto et al. [51]
Phase I
Selumetinib
MEKi + CT
Melosky et al. [44]
Phase II
Selumetinib
MEKi + CT
Hellmann et al. [56]
Phase Ib
Cobimetinib
MEKi + ICI
Gaudreau et al. [57]
Phase I/II
Trametinib
MEKi + ICI
Carter et al. [59]
Phase Ib
Selumetinib
MEKi + EGFR-TKI
Oxnard et al. [60]
Phase Ib
Selumetinib
MEKi + EGFR-TKI
MEKi, MEK inhibitors, BRAFi, BRAF inhibitors, CT, chemotherapy, ICI, immune checkpoint inhibitors, EGFR-TKI, epidermal growth factor receptor tyrosine kinase inhibitors
At the early stage, MEK inhibitors’ monotherapy had been detected a lot but seemed not to be effective for NSCLC patients for its poor efficacy and higher toxicities. No matter compared with pemetrexed or docetaxel, no significant difference in median PFS or OS was observed and dermatitis acneiform, hypertension, and diarrhea toxicities were more common [3639].
MEK inhibitors in combination with BRAF inhibitors as a treatment demonstrated an improved efficacy for NSCLC patients. Currently, trametinib combined with dabrafenib has been the only therapy approved by the United States Food and Drug Administration (FDA) and European Medicines Agency (EMA) for the treatment of BRAF V600E-mutant NSCLC patients, which has been written into the National Comprehensive Cancer Network (NCCN) guidelines as well. The phase II trials in previously treated and untreated BRAF V600E-mutant NSCLC demonstrated that PFS and OS were longer and ORR was higher, which were much better than the outcomes of single-agent BRAF inhibitor in the previous study [40, 41, 72]. According to these trials, NCCN guidelines recommend that dabrafenib combined with trametinib be the first-line and subsequent therapy for BRAF V600E mutation-positive NSCLC patients. However, the challenge with this combination is the emergence of drug resistance and no effective treatment strategy to overcome it yet. Another challenge in targeted therapy for non-V600E mutation patients is still lacking.
Chemotherapy plus MEK inhibitors have showed obscure clinical outcomes to date. Some trials demonstrated that this combination therapy had the trend of longer PFS, OS, and higher ORR, but with no significant difference, especially in the SELECT series trials [47, 49, 50]. Other trails [4446] tried to do some exploration in subgroup NSCLC patients, such as KRAS-mutant and KRAS-WT patients. Regrettably, different chemotherapy drugs seemed to influence the outcomes as well. A phase 1/1b study showed that ORR was 18% versus 24% in KRAS-WT and KRAS-mutant patients in trametinib plus docetaxel group, while ORR was 17% versus 11% KRAS-WT and KRAS-mutant patients in trametinib plus pemetrexed group [46]. Chemotherapy applied concurrently with MEK inhibitors requires further specific validation including the different chemotherapy agents, KRAS or other gene mutations and different MEK inhibitors before this combination strategy can become a standard treatment option for NSCLC patients.
Based on the preclinical studies, MEK inhibitors could improved T cell activation, conditioned the tumor microenvironment to facilitate improved response to anti-CTLA-4 treatment and prolonged survival in KRAS-mutant mice in combination with CTLA-4 blockade [54, 55]. However, the current relevant clinical trials of ICIs plus MEK inhibitors were not sufficient to draw the conclusion yet. Since only a phase Ib study [56] investigated the safety and efficacy of cobimetinib plus atezolizumab in a single arm for few NSCLC patients and several PD-1/L1 inhibitors plus MEK inhibitors clinical trials [57] (Table 4) are still ongoing, the finial clinical outcomes are worthy being looking forward to furthermore.
Although targeted therapy has dramatically changed our approach to treating NSCLC, the emergency of drug resistance and the lack of effective treatments to some special target such as KRAS still affect the prognosis of NSCLC patients. Preclinical data showed that MEK inhibitors plus EGFR-TKIs could inhibit cell proliferation significantly of EGFR-TKIs-resistant cells, while similar clinical trials have not been designed yet [58]. Current clinical trials [5962] focused on EGFR-TKIs, including erlotinib, osimertinib, and afatinib, in combination with MEK inhibitors appearing somewhat illusory for OS, PFS or ORR. These outcomes seemed not to be improved under this strategy and more obvious toxicities were revealed. Further researches should be designed more on the administration way to combine two or three drugs together to optimize the therapeutic effect in appropriate subset patients.
In addition to the emerging drugs and clinical studies mentioned above, there are still many more new treatment combinations that have conducted in early stages of clinical development. Novel combination drugs can be broadly classified as BRAF inhibitors, EGFR-TKIs, multi-target tyrosine kinase inhibitors, CDK4/6 inhibitors, ALK inhibitors, platin-based chemotherapy, and ICIs. Additionally, many treatment combinations being explored in early-stage clinical studies, such as PI3K and AKT inhibitors should be further detected in a more rational way with MEK inhibitors in human bodies [7377] (Fig. 2). The preclinical data indicated that the combined therapy of MEK and PI3K inhibitors has presented promising outcomes for NSCLC patients with the acquired resistance to EGFR-TKIs [78], but more clinical effects should be validated in the future. Overall, there seems to be hope on the horizon for NSCLC patients administrated with MEK inhibitors combined with other promising agents to improve patient outcomes finally.

Acknowledgements

Not applicable.
Not applicable.
Written informed consent for publication was obtained from all participants.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424.PubMed Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424.PubMed
2.
3.
Zurück zum Zitat Molina JR, Yang P, Cassivi SD, et al. Non-small cell lung cancer: epidemiology, risk factors, treatment, and survivorship. Mayo Clin Proc. 2008;83(5):584–94.PubMedCrossRef Molina JR, Yang P, Cassivi SD, et al. Non-small cell lung cancer: epidemiology, risk factors, treatment, and survivorship. Mayo Clin Proc. 2008;83(5):584–94.PubMedCrossRef
4.
Zurück zum Zitat N A Howlader N, Krapcho M, Miller D, Bishop K, Kosary Cl, Yu M, Ruhl J, Tatalovich Z, Mariotto A, Lewis DR, Chen HS, Feuer EJ, Cronin KA (Eds). SEER Cancer Statistics Review, 1975–2014. National Cancer Institute. 2016. N A Howlader N, Krapcho M, Miller D, Bishop K, Kosary Cl, Yu M, Ruhl J, Tatalovich Z, Mariotto A, Lewis DR, Chen HS, Feuer EJ, Cronin KA (Eds). SEER Cancer Statistics Review, 1975–2014. National Cancer Institute. 2016.
5.
Zurück zum Zitat Marchetti A, Felicioni L, Malatesta S, et al. Clinical features and outcome of patients with non-small-cell lung cancer harboring BRAF mutations. J Clin Oncol. 2011;29(26):3574–9.PubMedCrossRef Marchetti A, Felicioni L, Malatesta S, et al. Clinical features and outcome of patients with non-small-cell lung cancer harboring BRAF mutations. J Clin Oncol. 2011;29(26):3574–9.PubMedCrossRef
6.
Zurück zum Zitat Larkin J, Ascierto PA, Dreno B, et al. Combined vemurafenib and cobimetinib in BRAF-mutated melanoma. N Engl J Med. 2014;371(20):1867–76.PubMedCrossRef Larkin J, Ascierto PA, Dreno B, et al. Combined vemurafenib and cobimetinib in BRAF-mutated melanoma. N Engl J Med. 2014;371(20):1867–76.PubMedCrossRef
7.
Zurück zum Zitat Robert C, Karaszewska B, Schachter J, et al. Improved overall survival in melanoma with combined dabrafenib and trametinib. N Engl J Med. 2015;372(1):30–9.PubMedCrossRef Robert C, Karaszewska B, Schachter J, et al. Improved overall survival in melanoma with combined dabrafenib and trametinib. N Engl J Med. 2015;372(1):30–9.PubMedCrossRef
8.
Zurück zum Zitat Martinelli E, Morgillo F, Troiani T, et al. Cancer resistance to therapies against the EGFR-RAS-RAF pathway: the role of MEK. Cancer Treat Rev. 2017;53:61–9.PubMedCrossRef Martinelli E, Morgillo F, Troiani T, et al. Cancer resistance to therapies against the EGFR-RAS-RAF pathway: the role of MEK. Cancer Treat Rev. 2017;53:61–9.PubMedCrossRef
10.
Zurück zum Zitat Ricciuti B, Leonardi GC, Metro G, et al. Targeting the KRAS variant for treatment of non-small cell lung cancer: potential therapeutic applications. Expert Rev Respir Med. 2016;10(1):53–68.PubMedCrossRef Ricciuti B, Leonardi GC, Metro G, et al. Targeting the KRAS variant for treatment of non-small cell lung cancer: potential therapeutic applications. Expert Rev Respir Med. 2016;10(1):53–68.PubMedCrossRef
11.
Zurück zum Zitat Fischmann TO, Smith CK, Mayhood TW, et al. Crystal structures of MEK1 binary and ternary complexes with nucleotides and inhibitors. Biochemistry. 2009;48(12):2661–74.PubMedCrossRef Fischmann TO, Smith CK, Mayhood TW, et al. Crystal structures of MEK1 binary and ternary complexes with nucleotides and inhibitors. Biochemistry. 2009;48(12):2661–74.PubMedCrossRef
12.
Zurück zum Zitat Shaul YD, Seger R. The MEK/ERK cascade: from signaling specificity to diverse functions. Biochim Biophys Acta. 2007;1773(8):1213–26.PubMedCrossRef Shaul YD, Seger R. The MEK/ERK cascade: from signaling specificity to diverse functions. Biochim Biophys Acta. 2007;1773(8):1213–26.PubMedCrossRef
13.
Zurück zum Zitat Fukuda M, Gotoh Y, Nishida E. Interaction of MAP kinase with MAP kinase kinase: its possible role in the control of nucleocytoplasmic transport of MAP kinase. EMBO J. 1997;16(8):1901–8.PubMedPubMedCentralCrossRef Fukuda M, Gotoh Y, Nishida E. Interaction of MAP kinase with MAP kinase kinase: its possible role in the control of nucleocytoplasmic transport of MAP kinase. EMBO J. 1997;16(8):1901–8.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Takekawa M, Tatebayashi K, Saito H. Conserved docking site is essential for activation of mammalian MAP kinase kinases by specific MAP kinase kinase kinases. Mol Cell. 2005;18(3):295–306.PubMedCrossRef Takekawa M, Tatebayashi K, Saito H. Conserved docking site is essential for activation of mammalian MAP kinase kinases by specific MAP kinase kinase kinases. Mol Cell. 2005;18(3):295–306.PubMedCrossRef
15.
Zurück zum Zitat Guo YJ, Pan WW, Liu SB, et al. ERK/MAPK signalling pathway and tumorigenesis. Exp Ther Med. 2020;19(3):1997–2007.PubMedPubMedCentral Guo YJ, Pan WW, Liu SB, et al. ERK/MAPK signalling pathway and tumorigenesis. Exp Ther Med. 2020;19(3):1997–2007.PubMedPubMedCentral
16.
17.
Zurück zum Zitat Downward J. Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer. 2003;3(1):11–22.PubMedCrossRef Downward J. Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer. 2003;3(1):11–22.PubMedCrossRef
23.
Zurück zum Zitat Yeh TC, Marsh V, Bernat BA, et al. Biological characterization of ARRY-142886 (AZD6244), a potent, highly selective mitogen-activated protein kinase kinase 1/2 inhibitor. Clin Cancer Res. 2007;13(5):1576–83.PubMedCrossRef Yeh TC, Marsh V, Bernat BA, et al. Biological characterization of ARRY-142886 (AZD6244), a potent, highly selective mitogen-activated protein kinase kinase 1/2 inhibitor. Clin Cancer Res. 2007;13(5):1576–83.PubMedCrossRef
24.
Zurück zum Zitat Gilmartin AG, Bleam MR, Groy A, et al. GSK1120212 (JTP-74057) is an inhibitor of MEK activity and activation with favorable pharmacokinetic properties for sustained in vivo pathway inhibition. Clin Cancer Res. 2011;17(5):989–1000.PubMedCrossRef Gilmartin AG, Bleam MR, Groy A, et al. GSK1120212 (JTP-74057) is an inhibitor of MEK activity and activation with favorable pharmacokinetic properties for sustained in vivo pathway inhibition. Clin Cancer Res. 2011;17(5):989–1000.PubMedCrossRef
25.
Zurück zum Zitat Lee PA, Wallace E, Marlow A, et al. Abstract 2515: preclinical development of ARRY-162, a potent and selective MEK 1/2 inhibitor. Cancer Res 2014. Lee PA, Wallace E, Marlow A, et al. Abstract 2515: preclinical development of ARRY-162, a potent and selective MEK 1/2 inhibitor. Cancer Res 2014.
26.
Zurück zum Zitat Hoeflich KP, Merchant M, Orr C, et al. Intermittent administration of MEK inhibitor GDC-0973 plus PI3K inhibitor GDC-0941 triggers robust apoptosis and tumor growth inhibition. Cancer Res. 2012;72(1):210–9.PubMedCrossRef Hoeflich KP, Merchant M, Orr C, et al. Intermittent administration of MEK inhibitor GDC-0973 plus PI3K inhibitor GDC-0941 triggers robust apoptosis and tumor growth inhibition. Cancer Res. 2012;72(1):210–9.PubMedCrossRef
28.
Zurück zum Zitat Pheneger J, Wallace E, Marlow A, et al. Characterization of ARRY-438162, a potent MEK inhibitor in combination with methotrexate or ibuprofen in in vivo models of arthritis. In: Proceedings of the 2006 annual scientific meeting, Boston, MA, USA, 20–24 October 2006; p. 794. Pheneger J, Wallace E, Marlow A, et al. Characterization of ARRY-438162, a potent MEK inhibitor in combination with methotrexate or ibuprofen in in vivo models of arthritis. In: Proceedings of the 2006 annual scientific meeting, Boston, MA, USA, 20–24 October 2006; p. 794.
29.
Zurück zum Zitat Rice KD, Aay N, Anand NK, et al. Novel carboxamide-based allosteric MEK inhibitors: discovery and optimization efforts toward XL518 (GDC-0973). ACS Med Chem Lett. 2012;3(5):416–21.PubMedPubMedCentralCrossRef Rice KD, Aay N, Anand NK, et al. Novel carboxamide-based allosteric MEK inhibitors: discovery and optimization efforts toward XL518 (GDC-0973). ACS Med Chem Lett. 2012;3(5):416–21.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Inaba K, Oda K, Ikeda Y, et al. Antitumor activity of a combination of dual PI3K/mTOR inhibitor SAR245409 and selective MEK1/2 inhibitor pimasertib in endometrial carcinomas. Gynecol Oncol. 2015;138(2):323–31.PubMedCrossRef Inaba K, Oda K, Ikeda Y, et al. Antitumor activity of a combination of dual PI3K/mTOR inhibitor SAR245409 and selective MEK1/2 inhibitor pimasertib in endometrial carcinomas. Gynecol Oncol. 2015;138(2):323–31.PubMedCrossRef
31.
Zurück zum Zitat Ciuffreda L, Del Bufalo D, Desideri M, et al. Growth-inhibitory and antiangiogenic activity of the MEK inhibitor PD0325901 in malignant melanoma with or without BRAF mutations. Neoplasia. 2009;11(8):720–31.PubMedPubMedCentralCrossRef Ciuffreda L, Del Bufalo D, Desideri M, et al. Growth-inhibitory and antiangiogenic activity of the MEK inhibitor PD0325901 in malignant melanoma with or without BRAF mutations. Neoplasia. 2009;11(8):720–31.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Iverson C, Larson G, Lai C, et al. RDEA119/BAY 869766: a potent, selective, allosteric inhibitor of MEK1/2 for the treatment of cancer. Cancer Res. 2009;69(17):6839–47.PubMedCrossRef Iverson C, Larson G, Lai C, et al. RDEA119/BAY 869766: a potent, selective, allosteric inhibitor of MEK1/2 for the treatment of cancer. Cancer Res. 2009;69(17):6839–47.PubMedCrossRef
33.
Zurück zum Zitat Hatzivassiliou G, Haling JR, Chen H, et al. Mechanism of MEK inhibition determines efficacy in mutant KRAS- versus BRAF-driven cancers. Nature. 2013;501(7466):232–6.PubMedCrossRef Hatzivassiliou G, Haling JR, Chen H, et al. Mechanism of MEK inhibition determines efficacy in mutant KRAS- versus BRAF-driven cancers. Nature. 2013;501(7466):232–6.PubMedCrossRef
34.
Zurück zum Zitat Takahashi RH, Ma S, Robinson SJ, et al. Elucidating the mechanisms of formation for two unusual cytochrome P450-mediated fused ring metabolites of GDC-0623, a MAPK/ERK kinase inhibitor. Drug Metab Dispos. 2015;43(12):1929–33.PubMedCrossRef Takahashi RH, Ma S, Robinson SJ, et al. Elucidating the mechanisms of formation for two unusual cytochrome P450-mediated fused ring metabolites of GDC-0623, a MAPK/ERK kinase inhibitor. Drug Metab Dispos. 2015;43(12):1929–33.PubMedCrossRef
35.
Zurück zum Zitat Martinez-Garcia M, Banerji U, Albanell J, et al. First-in-human, phase I dose-escalation study of the safety, pharmacokinetics, and pharmacodynamics of RO5126766, a first-in-class dual MEK/RAF inhibitor in patients with solid tumors. Clin Cancer Res. 2012;18(17):4806–19.PubMedCrossRef Martinez-Garcia M, Banerji U, Albanell J, et al. First-in-human, phase I dose-escalation study of the safety, pharmacokinetics, and pharmacodynamics of RO5126766, a first-in-class dual MEK/RAF inhibitor in patients with solid tumors. Clin Cancer Res. 2012;18(17):4806–19.PubMedCrossRef
36.
Zurück zum Zitat Hainsworth JD, Cebotaru CL, Kanarev V, et al. 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(10):1630–6.PubMedCrossRef Hainsworth JD, Cebotaru CL, Kanarev V, et al. 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(10):1630–6.PubMedCrossRef
37.
Zurück zum Zitat Haura EB, Ricart AD, Larson TG, 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(8):2450–7.PubMedCrossRef Haura EB, Ricart AD, Larson TG, 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(8):2450–7.PubMedCrossRef
38.
Zurück zum Zitat Blumenschein GR Jr, Smit EF, Planchard D, et al. A randomized phase II study of the MEK1/MEK2 inhibitor trametinib (GSK1120212) compared with docetaxel in KRAS-mutant advanced non-small-cell lung cancer (NSCLC)†. Ann Oncol. 2015;26(5):894–901.PubMedPubMedCentralCrossRef Blumenschein GR Jr, Smit EF, Planchard D, et al. A randomized phase II study of the MEK1/MEK2 inhibitor trametinib (GSK1120212) compared with docetaxel in KRAS-mutant advanced non-small-cell lung cancer (NSCLC)†. Ann Oncol. 2015;26(5):894–901.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Lopez-Chavez A, Thomas A, Rajan A, et al. Molecular profiling and targeted therapy for advanced thoracic malignancies: a biomarker-derived, multiarm, multihistology phase II basket trial. J Clin Oncol. 2015;33(9):1000–7.PubMedPubMedCentralCrossRef Lopez-Chavez A, Thomas A, Rajan A, et al. Molecular profiling and targeted therapy for advanced thoracic malignancies: a biomarker-derived, multiarm, multihistology phase II basket trial. J Clin Oncol. 2015;33(9):1000–7.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Planchard D, Besse B, Groen HJM, et al. Dabrafenib plus trametinib in patients with previously treated BRAF(V600E)-mutant metastatic non-small cell lung cancer: an open-label, multicentre phase 2 trial. Lancet Oncol. 2016;17(7):984–93.PubMedPubMedCentralCrossRef Planchard D, Besse B, Groen HJM, et al. Dabrafenib plus trametinib in patients with previously treated BRAF(V600E)-mutant metastatic non-small cell lung cancer: an open-label, multicentre phase 2 trial. Lancet Oncol. 2016;17(7):984–93.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Planchard D, Smit EF, Groen HJM, et al. Dabrafenib plus trametinib in patients with previously untreated BRAF(V600E)-mutant metastatic non-small-cell lung cancer: an open-label, phase 2 trial. Lancet Oncol. 2017;18(10):1307–16.PubMedCrossRef Planchard D, Smit EF, Groen HJM, et al. Dabrafenib plus trametinib in patients with previously untreated BRAF(V600E)-mutant metastatic non-small-cell lung cancer: an open-label, phase 2 trial. Lancet Oncol. 2017;18(10):1307–16.PubMedCrossRef
42.
Zurück zum Zitat Salama AKS, Li S, Macrae ER, et al. Dabrafenib and trametinib in patients with tumors with BRAF(V600E) mutations: results of the NCI-MATCH trial subprotocol H. J Clin Oncol. 2020;38(33):3895–904.PubMedCrossRefPubMedCentral Salama AKS, Li S, Macrae ER, et al. Dabrafenib and trametinib in patients with tumors with BRAF(V600E) mutations: results of the NCI-MATCH trial subprotocol H. J Clin Oncol. 2020;38(33):3895–904.PubMedCrossRefPubMedCentral
43.
Zurück zum Zitat Jänne PA, Shaw AT, Pereira JR, et al. Selumetinib plus docetaxel for KRAS-mutant advanced non-small-cell lung cancer: a randomised, multicentre, placebo-controlled, phase 2 study. Lancet Oncol. 2013;14(1):38–47.PubMedCrossRef Jänne PA, Shaw AT, Pereira JR, et al. Selumetinib plus docetaxel for KRAS-mutant advanced non-small-cell lung cancer: a randomised, multicentre, placebo-controlled, phase 2 study. Lancet Oncol. 2013;14(1):38–47.PubMedCrossRef
44.
Zurück zum Zitat Melosky B, Bradbury P, Tu D, et al. Selumetinib in patients receiving standard pemetrexed and platinum-based chemotherapy for advanced or metastatic KRAS wildtype or unknown non-squamous non-small cell lung cancer: A randomized, multicenter, phase II study. Canadian Cancer Trials Group (CCTG) IND.219. Lung Cancer. 2019;133:48–55.PubMedCrossRef Melosky B, Bradbury P, Tu D, et al. Selumetinib in patients receiving standard pemetrexed and platinum-based chemotherapy for advanced or metastatic KRAS wildtype or unknown non-squamous non-small cell lung cancer: A randomized, multicenter, phase II study. Canadian Cancer Trials Group (CCTG) IND.219. Lung Cancer. 2019;133:48–55.PubMedCrossRef
45.
Zurück zum Zitat Jänne PA, Smith I, Mcwalter G, et al. Impact of KRAS codon subtypes from a randomised phase II trial of selumetinib plus docetaxel in KRAS mutant advanced non-small-cell lung cancer. Br J Cancer. 2015;113(2):199–203.PubMedPubMedCentralCrossRef Jänne PA, Smith I, Mcwalter G, et al. Impact of KRAS codon subtypes from a randomised phase II trial of selumetinib plus docetaxel in KRAS mutant advanced non-small-cell lung cancer. Br J Cancer. 2015;113(2):199–203.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Gandara DR, Leighl N, Delord JP, et al. A phase 1/1b study evaluating trametinib plus docetaxel or pemetrexed in patients with advanced non-small cell lung cancer. J Thorac Oncol. 2017;12(3):556–66.PubMedCrossRef Gandara DR, Leighl N, Delord JP, et al. A phase 1/1b study evaluating trametinib plus docetaxel or pemetrexed in patients with advanced non-small cell lung cancer. J Thorac Oncol. 2017;12(3):556–66.PubMedCrossRef
47.
Zurück zum Zitat Jänne PA, Van Den Heuvel MM, Barlesi F, et al. Selumetinib plus docetaxel compared with docetaxel alone and progression-free survival in patients with KRAS-mutant advanced non-small cell lung cancer: the SELECT-1 randomized clinical trial. JAMA. 2017;317(18):1844–53.PubMedPubMedCentralCrossRef Jänne PA, Van Den Heuvel MM, Barlesi F, et al. Selumetinib plus docetaxel compared with docetaxel alone and progression-free survival in patients with KRAS-mutant advanced non-small cell lung cancer: the SELECT-1 randomized clinical trial. JAMA. 2017;317(18):1844–53.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Kaufman J, Stinchcombe TE. Treatment of KRAS-mutant non-small cell lung cancer: the end of the beginning for targeted therapies. JAMA. 2017;317(18):1835–7.PubMedCrossRef Kaufman J, Stinchcombe TE. Treatment of KRAS-mutant non-small cell lung cancer: the end of the beginning for targeted therapies. JAMA. 2017;317(18):1835–7.PubMedCrossRef
49.
Zurück zum Zitat Soria JC, Fülöp A, Maciel C, et al. SELECT-2: a phase II, double-blind, randomized, placebo-controlled study to assess the efficacy of selumetinib plus docetaxel as a second-line treatment of patients with advanced or metastatic non-small-cell lung cancer. Ann Oncol. 2017;28(12):3028–36.PubMedPubMedCentralCrossRef Soria JC, Fülöp A, Maciel C, et al. SELECT-2: a phase II, double-blind, randomized, placebo-controlled study to assess the efficacy of selumetinib plus docetaxel as a second-line treatment of patients with advanced or metastatic non-small-cell lung cancer. Ann Oncol. 2017;28(12):3028–36.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Greystoke A, Steele N, Arkenau HT, et al. SELECT-3: a phase I study of selumetinib in combination with platinum-doublet chemotherapy for advanced NSCLC in the first-line setting. Br J Cancer. 2017;117(7):938–46.PubMedPubMedCentralCrossRef Greystoke A, Steele N, Arkenau HT, et al. SELECT-3: a phase I study of selumetinib in combination with platinum-doublet chemotherapy for advanced NSCLC in the first-line setting. Br J Cancer. 2017;117(7):938–46.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Seto T, Hirai F, Saka H, et al. Safety and tolerability of selumetinib as a monotherapy, or in combination with docetaxel as second-line therapy, in Japanese patients with advanced solid malignancies or non-small cell lung cancer. Jpn J Clin Oncol. 2018;48(1):31–42.PubMedCrossRef Seto T, Hirai F, Saka H, et al. Safety and tolerability of selumetinib as a monotherapy, or in combination with docetaxel as second-line therapy, in Japanese patients with advanced solid malignancies or non-small cell lung cancer. Jpn J Clin Oncol. 2018;48(1):31–42.PubMedCrossRef
52.
Zurück zum Zitat Rittmeyer A, Barlesi F, Waterkamp D, et al. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet. 2017;389(10066):255–65.CrossRefPubMed Rittmeyer A, Barlesi F, Waterkamp D, et al. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet. 2017;389(10066):255–65.CrossRefPubMed
53.
Zurück zum Zitat Herbst RS, Baas P, Kim DW, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet. 2016;387(10027):1540–50.CrossRefPubMed Herbst RS, Baas P, Kim DW, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet. 2016;387(10027):1540–50.CrossRefPubMed
54.
Zurück zum Zitat Choi H, Deng J, Li S, et al. Pulsatile MEK inhibition improves anti-tumor immunity and T cell function in murine Kras mutant lung cancer. Cell Rep. 2019;27(3):806-819.e805.PubMedPubMedCentralCrossRef Choi H, Deng J, Li S, et al. Pulsatile MEK inhibition improves anti-tumor immunity and T cell function in murine Kras mutant lung cancer. Cell Rep. 2019;27(3):806-819.e805.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Poon E, Mullins S, Watkins A, et al. The MEK inhibitor selumetinib complements CTLA-4 blockade by reprogramming the tumor immune microenvironment. J Immunother Cancer. 2017;5(1):63.PubMedPubMedCentralCrossRef Poon E, Mullins S, Watkins A, et al. The MEK inhibitor selumetinib complements CTLA-4 blockade by reprogramming the tumor immune microenvironment. J Immunother Cancer. 2017;5(1):63.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Hellmann MD, Kim TW, Lee CB, et al. Phase Ib study of atezolizumab combined with cobimetinib in patients with solid tumors. Ann Oncol. 2019;30(7):1134–42.PubMedPubMedCentralCrossRef Hellmann MD, Kim TW, Lee CB, et al. Phase Ib study of atezolizumab combined with cobimetinib in patients with solid tumors. Ann Oncol. 2019;30(7):1134–42.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Gaudreau PO, Lee JJ, Heymach JV, et al. 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(4):384–8.PubMedCrossRefPubMedCentral Gaudreau PO, Lee JJ, Heymach JV, et al. 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(4):384–8.PubMedCrossRefPubMedCentral
58.
Zurück zum Zitat Li S, Chen S, Jiang Y, et al. Synergistic interaction between MEK inhibitor and gefitinib in EGFR-TKI-resistant human lung cancer cells. Oncol Lett. 2015;10(4):2652–6.PubMedPubMedCentralCrossRef Li S, Chen S, Jiang Y, et al. Synergistic interaction between MEK inhibitor and gefitinib in EGFR-TKI-resistant human lung cancer cells. Oncol Lett. 2015;10(4):2652–6.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Carter CA, Rajan A, Keen C, et al. Selumetinib with and without erlotinib in KRAS mutant and KRAS wild-type advanced nonsmall-cell lung cancer. Ann Oncol. 2016;27(4):693–9.PubMedPubMedCentralCrossRef Carter CA, Rajan A, Keen C, et al. Selumetinib with and without erlotinib in KRAS mutant and KRAS wild-type advanced nonsmall-cell lung cancer. Ann Oncol. 2016;27(4):693–9.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Oxnard GR, Yang JC, Yu H, 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(4):507–16.PubMedCrossRef Oxnard GR, Yang JC, Yu H, 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(4):507–16.PubMedCrossRef
61.
Zurück zum Zitat Asco. Phase I study of afatinib plus selumetinib in patients with KRAS mutation-positive colorectal, non-small cell lung and pancreatic cancer. 2020. Asco. Phase I study of afatinib plus selumetinib in patients with KRAS mutation-positive colorectal, non-small cell lung and pancreatic cancer. 2020.
62.
Zurück zum Zitat Esmo. Phase I study of lapatinib and trametinib in patients with KRAS mutant colorectal, non-small cell lung and pancreatic cancer. 2019. Esmo. Phase I study of lapatinib and trametinib in patients with KRAS mutant colorectal, non-small cell lung and pancreatic cancer. 2019.
63.
Zurück zum Zitat Villanueva J, Infante JR, Krepler C, et al. Concurrent MEK2 mutation and BRAF amplification confer resistance to BRAF and MEK inhibitors in melanoma. Cell Rep. 2013;4(6):1090–9.PubMedPubMedCentralCrossRef Villanueva J, Infante JR, Krepler C, et al. Concurrent MEK2 mutation and BRAF amplification confer resistance to BRAF and MEK inhibitors in melanoma. Cell Rep. 2013;4(6):1090–9.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Emery CM, Vijayendran KG, Zipser MC, et al. MEK1 mutations confer resistance to MEK and B-RAF inhibition. Proc Natl Acad Sci U S A. 2009;106(48):20411–6.PubMedPubMedCentralCrossRef Emery CM, Vijayendran KG, Zipser MC, et al. MEK1 mutations confer resistance to MEK and B-RAF inhibition. Proc Natl Acad Sci U S A. 2009;106(48):20411–6.PubMedPubMedCentralCrossRef
65.
Zurück zum Zitat Ciuffreda L, McCubrey JA, Milella M. Signaling intermediates (PI3K/PTEN/AKT/mTOR and RAF/MEK/ERK Pathways) as therapeutic targets for anti-cancer and anti-angiogenesis treatments. Curr Signal Transduct Ther. 2009;4(2):130–43.CrossRef Ciuffreda L, McCubrey JA, Milella M. Signaling intermediates (PI3K/PTEN/AKT/mTOR and RAF/MEK/ERK Pathways) as therapeutic targets for anti-cancer and anti-angiogenesis treatments. Curr Signal Transduct Ther. 2009;4(2):130–43.CrossRef
66.
Zurück zum Zitat Yao W, Yue P, Zhang G, et al. Enhancing therapeutic efficacy of the MEK inhibitor, MEK162, by blocking autophagy or inhibiting PI3K/Akt signaling in human lung cancer cells. Cancer Lett. 2015;364(1):70–8.PubMedPubMedCentralCrossRef Yao W, Yue P, Zhang G, et al. Enhancing therapeutic efficacy of the MEK inhibitor, MEK162, by blocking autophagy or inhibiting PI3K/Akt signaling in human lung cancer cells. Cancer Lett. 2015;364(1):70–8.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Meng J, Dai B, Fang B, et al. Combination treatment with MEK and AKT inhibitors is more effective than each drug alone in human non-small cell lung cancer in vitro and in vivo. PLoS ONE. 2010;5(11):e14124.PubMedPubMedCentralCrossRef Meng J, Dai B, Fang B, et al. Combination treatment with MEK and AKT inhibitors is more effective than each drug alone in human non-small cell lung cancer in vitro and in vivo. PLoS ONE. 2010;5(11):e14124.PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Engelman JA, Chen L, Tan X, et al. Effective use of PI3K and MEK inhibitors to treat mutant Kras G12D and PIK3CA H1047R murine lung cancers. Nat Med. 2008;14(12):1351–6.PubMedPubMedCentralCrossRef Engelman JA, Chen L, Tan X, et al. Effective use of PI3K and MEK inhibitors to treat mutant Kras G12D and PIK3CA H1047R murine lung cancers. Nat Med. 2008;14(12):1351–6.PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Li H, Schmid-Bindert G, Wang D, et al. Blocking the PI3K/AKT and MEK/ERK signaling pathways can overcome gefitinib-resistance in non-small cell lung cancer cell lines. Adv Med Sci. 2011;56(2):275–84.PubMedCrossRef Li H, Schmid-Bindert G, Wang D, et al. Blocking the PI3K/AKT and MEK/ERK signaling pathways can overcome gefitinib-resistance in non-small cell lung cancer cell lines. Adv Med Sci. 2011;56(2):275–84.PubMedCrossRef
70.
Zurück zum Zitat Falcone I, Conciatori F, Bazzichetto C, et al. Tumor microenvironment: implications in melanoma resistance to targeted therapy and immunotherapy. Cancers (Basel). 2020;12(10):2870.CrossRef Falcone I, Conciatori F, Bazzichetto C, et al. Tumor microenvironment: implications in melanoma resistance to targeted therapy and immunotherapy. Cancers (Basel). 2020;12(10):2870.CrossRef
71.
Zurück zum Zitat Qu Y, Wu X, Yin Y, et al. Antitumor activity of selective MEK1/2 inhibitor AZD6244 in combination with PI3K/mTOR inhibitor BEZ235 in gefitinib-resistant NSCLC xenograft models. J Exp Clin Cancer Res. 2014;33:52.PubMedPubMedCentralCrossRef Qu Y, Wu X, Yin Y, et al. Antitumor activity of selective MEK1/2 inhibitor AZD6244 in combination with PI3K/mTOR inhibitor BEZ235 in gefitinib-resistant NSCLC xenograft models. J Exp Clin Cancer Res. 2014;33:52.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Planchard D, Kim TM, Mazieres J, et al. Dabrafenib in patients with BRAF(V600E)-positive advanced non-small-cell lung cancer: a single-arm, multicentre, open-label, phase 2 trial. Lancet Oncol. 2016;17(5):642–50.PubMedPubMedCentralCrossRef Planchard D, Kim TM, Mazieres J, et al. Dabrafenib in patients with BRAF(V600E)-positive advanced non-small-cell lung cancer: a single-arm, multicentre, open-label, phase 2 trial. Lancet Oncol. 2016;17(5):642–50.PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat Tolcher AW, Kurzrock R, Valero V, et al. Phase I dose-escalation trial of the oral AKT inhibitor uprosertib in combination with the oral MEK1/MEK2 inhibitor trametinib in patients with solid tumors. Cancer Chemother Pharmacol. 2020;85(4):673–83.PubMedCrossRef Tolcher AW, Kurzrock R, Valero V, et al. Phase I dose-escalation trial of the oral AKT inhibitor uprosertib in combination with the oral MEK1/MEK2 inhibitor trametinib in patients with solid tumors. Cancer Chemother Pharmacol. 2020;85(4):673–83.PubMedCrossRef
74.
Zurück zum Zitat Shapiro GI, Lorusso P, Cho DC, et al. A phase Ib open-label dose escalation study of the safety, pharmacokinetics, and pharmacodynamics of cobimetinib (GDC-0973) and ipatasertib (GDC-0068) in patients with locally advanced or metastatic solid tumors. Invest New Drugs 2020. Shapiro GI, Lorusso P, Cho DC, et al. A phase Ib open-label dose escalation study of the safety, pharmacokinetics, and pharmacodynamics of cobimetinib (GDC-0973) and ipatasertib (GDC-0068) in patients with locally advanced or metastatic solid tumors. Invest New Drugs 2020.
75.
Zurück zum Zitat Bardia A, Gounder M, Rodon J, 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. Oncologist. 2020;25(1):e160–9.PubMedCrossRef Bardia A, Gounder M, Rodon J, 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. Oncologist. 2020;25(1):e160–9.PubMedCrossRef
76.
Zurück zum Zitat Shapiro GI, Lorusso P, Kwak E, et al. Phase Ib study of the MEK inhibitor cobimetinib (GDC-0973) in combination with the PI3K inhibitor pictilisib (GDC-0941) in patients with advanced solid tumors. Invest New Drugs. 2020;38(2):419–32.PubMedCrossRef Shapiro GI, Lorusso P, Kwak E, et al. Phase Ib study of the MEK inhibitor cobimetinib (GDC-0973) in combination with the PI3K inhibitor pictilisib (GDC-0941) in patients with advanced solid tumors. Invest New Drugs. 2020;38(2):419–32.PubMedCrossRef
77.
Zurück zum Zitat Ramanathan RK, Von Hoff DD, Eskens F, et al. Phase Ib trial of the PI3K inhibitor copanlisib combined with the allosteric MEK inhibitor refametinib in patients with advanced cancer. Target Oncol. 2020;15(2):163–74.PubMedPubMedCentralCrossRef Ramanathan RK, Von Hoff DD, Eskens F, et al. Phase Ib trial of the PI3K inhibitor copanlisib combined with the allosteric MEK inhibitor refametinib in patients with advanced cancer. Target Oncol. 2020;15(2):163–74.PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat Sato H, Yamamoto H, Sakaguchi M, et al. Combined inhibition of MEK and PI3K pathways overcomes acquired resistance to EGFR-TKIs in non-small cell lung cancer. Cancer Sci. 2018;109(10):3183–96.PubMedPubMedCentralCrossRef Sato H, Yamamoto H, Sakaguchi M, et al. Combined inhibition of MEK and PI3K pathways overcomes acquired resistance to EGFR-TKIs in non-small cell lung cancer. Cancer Sci. 2018;109(10):3183–96.PubMedPubMedCentralCrossRef
Metadaten
Titel
MEK inhibitors for the treatment of non-small cell lung cancer
verfasst von
Jing Han
Yang Liu
Sen Yang
Xuan Wu
Hongle Li
Qiming Wang
Publikationsdatum
01.12.2021
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2021
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-020-01025-7

Weitere Artikel der Ausgabe 1/2021

Journal of Hematology & Oncology 1/2021 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.