Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Feb 6.
Published in final edited form as: Curr Oncol Rep. 2021 Feb 6;23(2):23. doi: 10.1007/s11912-020-01008-4

Tumor and Systemic Immunomodulatory Effects of MEK Inhibition

Lauren Dennison 1, Aditya A Mohan 1, Mark Yarchoan 1
PMCID: PMC8028056  NIHMSID: NIHMS1682739  PMID: 33547983

Abstract

Purpose of Review

Mitogen-activated protein kinase (MAPK) kinase (MEK) is an integral component of the RAS signaling pathway, one of the most frequently mutated pathways in cancer biology. MEK inhibitors were initially developed to directly target oncogenic signaling, but are recognized to have pleiotropic effects on both tumor cells and lymphocytes. Here we review the preclinical and clinical evidence that MEK inhibition is immunomodulatory and discuss the potential rationale for combining MEK inhibitors with systemic immunotherapies.

Recent Findings

MEK inhibition may modulate the tumor microenvironment (TME) through direct effects on both tumor cells and immune cells. Despite encouraging evidence that MEK inhibition can reprogram the tumor microenvironment (TME) and augment anti-tumor immunity regardless of KRAS/BRAF status, recent clinical outcome studies combining MEK inhibition with systemic immunotherapy have yielded mixed results. The combination of MEK inhibitors plus systemic immunotherapies has been tolerable, but has thus far failed to demonstrate clear evidence of synergistic clinical activity. These results underscore the need to understand the appropriate therapeutic context for this combination.

Summary

MEK inhibitors have the potential to inhibit oncogenic signaling and reprogram the tumor immune microenvironment, representing an attractive therapy to combine with systemic immunotherapies. Ongoing preclinical and clinical studies will further clarify the immunomodulatory effects of MEK inhibitors to inform the design of rational therapeutic combinations.

Keywords: MEK, MAPK, PD-1, PD-L1, immunotherapy, RAS

Introduction

The MAPK pathway is a highly conserved signal transduction pathway in all eukaryotic cells that regulates a variety of normal cellular functions, including cell proliferation, differentiation, survival, and apoptosis (1). Upon activation of upstream growth factor receptors, RAS, the principal upstream activator of the MAPK pathway, undergoes a conformation change in which guanosine diphosphate (GDP) is exchanged for guanosine triphosphate (GTP)(2)’(3). Genes encoding RAS (KRAS, NRAS, HRAS) are among the most frequently mutated genes in a variety of cancer, including pancreas (90%), colon (50%), thyroid (50%), lung (30%) and melanoma (25%)(4). The conformational change of RAS results in the recruitment and activation of the RAF family of serine-threonine protein kinases (RAF-1, B-RAF, and A-RAF), which activate MEK1/2 (MKK or MAPK kinase) by phosphorylation. MEK1/2 then activates ERK1/2, the canonical mitogen-activated protein kinase (MAPK), via phosphorylation(3). Activated MAPKs can go on to phosphorylate a range of downstream substrates, including protein kinases and transcription factors, which, in turn, lead to the transcription of MAPK-regulated genes. In addition to transcriptional regulation, MAPKs can also regulate gene expression of targets by altering mRNA stability, transport, and translation (Figure 1)(2)’ (5).

Figure 1.

Figure 1.

Summary of the immunomodulatory effects of MEK inhibition on tumor cells and immune cell populations.

MEK is considered a central component in the MAPK signaling pathway, and MEK1/2 can be activated by several kinases including Mos, A-Raf, B-Raf, Raf-1, and MEKK (6). However, MEK1 and MEK2 are the only known regulators of ERK1/2, and thus the signals of these upstream activators converge at the level of MEK. Since ERK1/2 have been shown to regulate hundreds of downstream targets, MEK1/2 are seen as “gatekeepers” in the MAPK pathway. Therefore, MEK inhibition (MEKi) represents an attractive drug candidate for tumors that are addicted to the RAS-signaling pathway. Since the first synthetic small-molecule inhibitor of MEKI and MEK2 kinase activity was described in 1995, four small-molecule MEK inhibitors have been approved by the FDA for clinical use in various settings—trametinib, cobimetinib, binimetinib, and selumetinib — and multiple other MEK inhibitors are in clinical or preclinical stages of development (7),(8).

Although MEK inhibitors were developed to target oncogenic signaling, there is an increasing interest in understanding the immune consequences of systemic MEK inhibition. In the current era in which immunotherapy has become firmly established as a pillar of cancer therapy, some patients may receive both targeted inhibition of MEK and immune checkpoint immunotherapy throughout their cancer treatment. In certain tumor types for which MEK inhibition and immune checkpoint inhibitors have substantial efficacy (for example, BRAF mutated melanoma or BRAF mutated colorectal cancer with mismatch repair deficiency), understanding the immune consequences of MEK inhibition may have significant implications for the sequencing of cancer therapies, and also for determining whether these therapies can rationally be combined (9-12). Although targeted inhibition of RAS signaling in such tumors can have rapid and substantial anti-tumor activity, these effects are rarely durable due to the emergence of drug-resistant clones. By contrast, novel immune checkpoint inhibitors have the potential to transform short-lived responses into durable responses, providing a foundation for studying these therapies in combination.

The effects of MAPK signaling and modulation on cancer immunotherapy are complex and context-dependent. In addition to serving as a critical regulator of cancer growth and metastasis, the mitogen-activated protein kinases (MAPK) pathway is also involved in nearly all aspects of immune responses: from the initiation phase of innate immunity, to activation of adaptive immunity, to cell death to prevent uncontrolled immune activation (5). Multiple preclinical studies have found that MEK inhibitors have immunomodulatory effects, including increased tumor immunogenicity and the potential for synergy with systemic immune checkpoint inhibition. However, direct clinical evidence that MEK inhibitors can synergize with systemic immunotherapies is lacking, and the combination of a MEK inhibitor and a PD-L1 inhibitor recently failed to show compelling efficacy in a pivotal clinical trial of colorectal cancer (IMblaze370), a tumor type for which MEK inhibition and immune checkpoint therapy have no meaningful activity in unselected patients (13). Herein we summarize the complex and at times, contradictory effects of MEK inhibitors on the relationship between the tumor and immune environment to better elucidate rational drug combinations.

MAPK Pathway Effects Tumor Cell Immunogenicity

MEK signaling is a key pathway involved in both tumor cell survival and lymphocyte response to antigen stimulation and may modulate the TME through direct effects on both tumor cells and immune cells. Here we summarize the known effects of MEKi on tumor cells that may impact tumor immunogenicity, including alterations in Major histocompatibility complex class I (MHC- I) expression and antigen presentation on target cells, programmed death ligand 1 (PDL-1) expression, and changes in cytokine release.

MHC-I Expression

MHC-I molecules present short peptides from either foreign or native intracellular proteins on the cell surface. The recognition of cancer neoantigens (abnormal proteins not found on normal host cells) in the context of MHC by cytotoxic T cells is a critical means of cancer elimination by the host immune system (14,15). Downregulation of MHC class I molecules by cancer cells results in reduced sensitivity to lysis by cytotoxic T cells and NK cells, providing a mechanism for immune evasion. Consequently, tumors with low-level expression of MHC-I are associated with a poor prognosis (16,17). In order to identify negative regulators of MHC-I, Brea et al. used a pooled human kinome shRNA interference-based approach and identified MAP2K1 (MEK) as a negative regulator of MHC-I expression (18). This observed negative regulation of the MAPK pathway has been confirmed across many studies and cancer cell lines, in which treatment with different MEK inhibitors consistently led to increased MHC-I expression (7),(19),(20), (21),(22). This increase in MHC-I expression is shown to be associated with the upregulation of components of the antigen presentation pathway, such as B2M, TAP1, and TAP2 at both the mRNA and protein level (18),(21). Importantly, this increase in MHC-I expression on the cell surface enhanced the presentation of antigens, resulting in an increase in specific T-cell activity, confirming the important functional significance (22), (18), (21), (23).

While many have shown that MEK inhibition leads to an increase in MHC-I, it is of interest that the use of an EGFR inhibitor also resulted in the upregulation of surface MHC-I in an NSCLC cell line with activating mutations in EGFR and NRAS, despite no change in pERK levels. This suggests potential activation of a parallel pathway that regulates MHC-I expression in EGFR mutant cancers and implies that the MAPK pathway is not solely responsible for the regulation of surface MHC-I (18).

PD-L1 Expression

PD-L1 (B7-H1, CD274) is a transmembrane protein that has received recent attention for its role in suppressing the immune system. Binding of PD-L1 to its receptor, programmed death receptor (PD-1), found on activated T cells, B cells, and myeloid cells, leads to suppression of T-cell activation and proliferative cytokine release and induction of T-cell apoptosis (24,25). PD-L1 expression is induced on antigen presenting cells by interferons, released by T cells upon recognition of their cognate antigens, and serves as a mechanism to control the level of immune response. However, it is now recognized that tumor cells can co-opt this checkpoint pathway by expressing PD-L1, facilitating immune evasion (26). Unsurprisingly, PD-L1 overexpression is correlated with poor overall survival and progression-free survival in several solid tumor types, including gastric, urothelial, hepatocellular carcinoma, breast, and melanoma. Consequently, there is increased potential for benefit from α-PD1 or PD-L1 immunotherapy (27).

While is seems undisputed that the MAPK pathway is a negative regulator of MHC-I, the effects of pathway activation and inhibition on PD-L1 expression are less clear and may vary by cancer type. Mechanistically, it was shown that RAS can drive cell-intrinsic PD-L1 expression via a downstream signaling cascade (28). However, the idea that activated RAS leads directly to increased PD-L1 is likely oversimplified as many other mutations and microenvironments can influence MAPK cascade cross-talk (29). In support of this notion, several studies have seen no impact on PD-L1 expression in various cancer cells lines or transient increases that didn’t track with pathway activation upon treatment with MEKis(19), (30). This suggests an uncoupling of MAPK pathway activation levels and PD-L1 expression in cancer cells continually exposed to MEK inhibitors. Other studies have reported variable responses across cell lines, with both increases and decreases seen depending on the cell line and experimental context, though the effect was independent of alterations in the MAPK pathway (30)’(31). These conflictions may explain the lack of consistent significant clinical synergy seen between MEK inhibitors and α-PD1 or PD-L1 immune-checkpoint inhibitors and suggests the need to better understand the impact of the genetic background on tumor PD-L1 regulation (29).

Cytokine release

Tumors secrete various factors such as VEGF, IL-6, and IL-10 that are critical cytokines in the tumor microenvironment, playing a role in immune suppression (32). Collectively, these cytokines have been shown to promote the accumulation of a heterogeneous population of immunosuppressive cells including tumor-associated macrophages (TAMs), myeloid derived suppressor cells (MDSCs) or immature dendritic cells. These cell populations have been shown to inhibit anti-tumor immunity through a variety of mechanisms, such as depletion of arginine, recruitment of T-regulatory cells, and increasing reactive oxygen species (33).

Melanoma cell lines with constitutive MAPK pathway activation showed high levels of VEGF, IL-10, and IL-6 expression (34). Use of the MEKi U0126 resulted in decreased mRNA and protein levels of these soluble immunosuppressive factors. Importantly, this decrease was associated with an increase in the production of the inflammatory cytokines IL-12 and TNFα by dendritic cells. Additionally, KRAS mutated cells were shown to produce increased osteopontin, a cytokine that has been shown to attract TAMs and drive the expansion of the suppressive MDSC population in the spleen (35,36). Treatment of tumor cells with trametinib reduced the production of osteopontin, abrogating the cytokine driven expansion of mMDSCs from myeloid progenitors (37).

MEK Inhibition Effects Immune Cell Populations

T cells

The MAPK axis plays a significant role in T-cell function. Upon engagement of the T-cell receptor, the RAS-MAPK signaling pathway is activated, leading to proliferation and cytotoxic effector functions against tumor cells (38). Consequently, there are concerns that MEK inhibition may dampen T-cell anti-tumor immunity by inhibiting T-cell receptor signaling. In support of this notion, the MEK inhibitor UO126 was shown to have deleterious effects on T-cell proliferation in vitro and IFN- γ production by T cells when antigen-specific T cells were cultured with melanoma cells (39)(40). This negative effect on T-cell proliferation in vitro was seen in multiple T-cell subsets including naïve, effector, and memory (41)(42). The decrease in T- cell proliferation in vitro has been partially attributed to the suppression of the production of growth-promoting cytokines IL-2, TNFα, and IL-3 by stimulated T cells, and exogenous addition of IL-2 reversed the phenotype (43). Importantly, the suppressive effects of MEK inhibitors seen on T-cells were not limited to in vitro models. In an ovarian cancer mouse model, antigen-specific CD8 T cell were shown to have decreased proliferative capacity after treatment with trametinib (41). Additionally, IFN-γ production by CD8+ and CD4+ T cells isolated from mouse mammary carcinomas treated daily with trametinib was measured and a significant reduction was observed (20).

Other studies found that in vitro results that show decreased functional T-cell activity with MEK inhibitors did not fully translate in vivo. Treatment of established CT26 tumors with trametinib increased tumor CD4+ T cell proliferation and infiltration in terms of absolute quantification, although it should be noted that the Treg population was not distinguished from the helper T-cells. Tregs have been shown to have a decreased functional dependence on the MAPK pathway allowing suppressive capabilities to be spared with MEK inhibition (30), (42). Hu-Lieskovan et al. found that while MEKi impaired production of IFN-γ by splenocytes treated in culture, in vivo cytokine production was not effected following isolation from trametinib treated mice and ex vivo re-stimulation (44). In this study, however, trametinib was co-administered with high doses of IL-2. It is possible that this is a confounding variable, as one of the reasons cited for decreased T cell proliferation and IFN-γ production with MEK inhibition was decreased IL-2 production that was reversible upon exogenous IL-2 addition (43). Finally, in a CT26 tumor model, naïve CD8+ T-cell priming was impaired in the lymph nodes after treatment, but interestingly, an increase of antigen-specific CD8+ T cells was seen in the tumor after initiation of treatment relative to control mice (45). These tumor-infiltrating CD8+ T cells were shown to be protected from death driven by chronic TCR stimulation and maintained cytotoxic activity, although once again exogenous IL-2 was added when cytotoxic activity was measured. This apparent increase and improved effector phenotype of CD8+ T cells did not have a functional impact however, as depleting these CD8+ T cells in MEKi treated tumors did not affect tumor growth (45).

In summary, studies tend to agree that MEK inhibition significantly hinders T-cell function and proliferation in vitro. While this finding was translated in vivo in a number of studies, the inconsistencies seen in other studies may be further explained by the time points at which tumor and TILs were analyzed and the treatment schedule, in addition to the differences in tumor models which further influence immune cell populations. This suggests that future studies should focus on finding a treatment schedule that maximizes the positive effects seen.

Immunosuppressive populations

Neoplastic growth can result in a chronic inflammatory response that promotes the expansion and recruitment of myeloid-derived suppressor cells (MDSCs) and tumor associated macrophages (TAM). These heterogeneous and plastic populations hinder anti-tumor immunity by blocking T-cell functions and proliferation through the production of iNOS and arginase and directly drive tumor growth by promoting cancer sternness, angiogenesis, and metastasis formation (46,47). Several studies had shown a reduction in monocytic-MDSCs (mMDSCs) upon treatment with MEK inhibitors (37)(48). This was accompanied by a decrease in arginase production, an indirect explanation for the increase of tumor CD8+ T cells seen in some studies, suggesting that the final effect of MEKi on T-cell numbers is dependent on many different pieces (37). In addition to impaired MDSC recruitment, MEK inhibition has been shown to inhibits monocyte to TAM differentiation, thereby preventing the accumulation of TAMs (49).

B cells

While new evidence has pointed to the importance of B cells in modulating the immune response to tumors, very few studies have looked at how targeted therapy, and in particular MEK inhibition, impacts B-cell populations (50). B cells are typically characterized by their positive modulation of immune responses and inflammation achieved through antibody production as well as their ability to promote T-cell activation via antigen presentation (51). Increasingly, more studies are reporting on the existence of discrete subsets of B-regulator cells (52,53). These immunosuppressive cells are able to maintain immune tolerance, suppressing responses to neoplastic growth through the release of anti-inflammatory mediators such as IL-10 and the expression of PD-L1, as well as facilitating the conversion of T cells to T regulatory cells (50). A recent study from our group using a colorectal cancer mouse model found that MEK inhibitors can enhance anti-tumor immunity through the reprogramming of B cells, resulting in a reduced number of Bregs in the tumor and non-tumor draining lymph nodes (54).

The effects of MEK inhibition on immune cell populations are summarized in Figure 1.

MEK Inhibition and Immunotherapy

Blocking antibodies

Immune checkpoint inhibitors represent one of the most promising approaches in the treatment of cancer (55,56). As mentioned earlier, the ability of cancer cells to overcome immunosurveillance is a key factor leading to cancer progression. Drugs interrupting immune checkpoints, such as anti-CTLA-4, anti-PD-1, anti-PD-L1, and others that are in early development, have the potential to unleash anti-tumor immunity, but the percentage of patients that have shown durable responses to immunotherapy is small (26), (57). As a result, there is a growing interest in combining immunotherapy agents with a second therapeutic agent to maximize anti-tumor outcome (58). While immunotherapies and targeted therapies have distinctly different mechanisms of action, it is thought that combinations of both modalities may create synergies with increased benefit for cancer patients.

Consistently across tumor models, the combination of MEK inhibition with immune checkpoint inhibitors resulted in reduced tumor volume and increased overall survival (19),(30),(49),(45). This was frequently accompanied by an increase in tumor CD8+ and CD4+ T-cell number and proliferative capacity as well as increased expression of IFNγ and Granzyme B (GZMB). MEK inhibition also reversed the increase in Cox2 and Arg1 expression in tumors seen with anti-CTLA4 treatment (49). However, of note the sequence of dosing has been shown to impact in vivo efficacy, with the most potent results observed when the MEK inhibitor was given first (30), (49). This suggests that blocking MAPK signaling is important to prime and synergize tumors in response to immunotherapy through mechanisms explained above such as upregulation of MHC molecules and reduction of immunosuppression factors.

Agonist antibodies

Costimulatory receptor signaling plays an important role in regulating antigen-specific T-cell proliferation, differentiation, and apoptosis (59,60). OX40 (CD134) and 4-1BB (CD137) belong to the tumor necrosis factor (TNF) receptor (TNFR) family and represent T-cell costimulatory receptors expressed following primary activation via antigen-specific receptors and CD28 costimulatory receptors. The ligation of OX40 with OX40 ligand (OX40L), expressed on antigen presenting cells, leads to improved T-cell mediated anti-tumor immunity, with augmented CD4+ and CD8+ T-cell clonal expansion, effector differentiation, and survival, and in some cases abrogated suppressive activity of T-regs (61). Activation of the 4-1BB/4-1BBL–mediated signaling cascade has been shown to play a role in T-cell proliferation, activation-induced cell death prevention, eradication of established tumors, enhancement of integrin-mediated cell adherence, and amplification of cytotoxic t-cell responses (62,63). In order to overcome the potentially detrimental effects of MEK inhibition on T-cell function there is a growing interest in combination therapy with agonist antibodies. Importantly, these antibodies have been shown to activate T-cells independently of MEK1/2 signaling, theoretically allowing the restoration of effector function even in the presence of MEK inhibitors (64).

To test this notion, Dushyanthen et al. investigated the benefit of combining agonist therapies with the MEK inhibitor trametinib (20). They found that while MEK inhibition increased tumor immunogenicity, it significantly inhibited early T-cell signaling, reducing both the proliferation and IFN-γ production of CD8+ and CD4+ T-cells in MAPK/Ras driven TNBC tumors. Both α-OX-40 Ab and α-4-1BB Ab restored the long-term proliferation and cytokine production of T-cell populations both in vitro and in vivo, confirming the ability of these agonists to prevent the impairment of T-cell function. Additionally, triple combination treatment with Trametinib, α-PD-1, and α- OX-40 Ab/α-4-1BB Ab resulted in significantly enhanced inhibition of tumor growth and prolonged survival. Of interest, anti-4-1BB/anti-OX-40 monotherapy enhanced the frequency of MDSCs, which was reduced following the addition of Trametinib (20). While these results are exciting, more studies need to be done in additional models to confirm how broadly these findings can be applied.

Clinical Trials

Over the past decade, immune checkpoint inhibitors have revolutionized the treatment of many cancers and have become firmly established as a pillar of cancer therapy. However, it is currently estimated that only approximately 40% of cancers are candidates for treatment with immune checkpoint inhibitors, and of these tumors only approximately 15% will respond (65). Novel immunotherapy combinations that can extend the benefit of immune checkpoint inhibitors to immune-resistant tumor types remains a critically important area of discovery. In principle, beyond the potential for immunomodulation, the combination of targeted therapies with systemic immunotherapies may be rational. Targeted therapies such as MEK inhibitors can induce rapid and significant tumor regressions, but these responses are often transient because of acquired resistance mutations. The addition of immune checkpoint inhibitors may in theory facilitate tumor elimination, converting a transient clinical response into a more durable responses. The success of combination therapy with a MEK inhibitor and immune checkpoint inhibitors in preclinical models led to the clinical investigation of such combinations in a number of different clinical contexts (Table 1). Despite initial excitement, the results of these trials combining MEK inhibitors with systemic immunotherapies thus far generally failed to show convincing evidence of synergistic antitumor activity.

Table 1.

Clinical trials investigating the combination of MEK inhibitors with Immunomodulatory Agents.

MEK Inhibitor Indication Design Phase # of patients Results NCT Reference
Cobimetinib Advanced BRAFV600 Wild-type Melanoma Cobimetinib and Atezolizumab vs Pembrolizumab; Randomized III 450 No improvement in Progression Free Survival NCT03273153 Miller WH, Kim TM, Lee CB, et al. J Clin Oncol. 2017;35(suppl; abstr 3057). doi: 10.1200/JCO.2017.35.15_suppl.3057
Gallbladder Carcinoma, Stage IV intrahepatic Cholangiocarcinom a, unresectable Cholangiocarcinom a Atezolizumab vs Atezolizumab and Cobimetinib; Randomized II 82 Improvement in Progression Free Survival and Disease Control Rate. NCT03201458 N/A
Breast Cancer Cobimetinib+Paclitaxel vs Cobimetinib+Atezolizumab+Paclitaxel vs Cobimetinib+Atezolizumab+Nab-Paclitaxel; Randomized II 169 ORRs were similar between the A + C + P arm and A + C + nP arm. Numerically higher ORR and PFS were observed in pts with PD-L1+ disease NCT02322814 Brufesky A, Kim SB, Zvirbule Z, et al. Journal of Clinical Oncology 37, no 15_suppl (May 20, 2019)1013-1013. doi:10.1200/JCO.2019.37.15_suppl.1013
Colorectal Cancer ·Atezolizumab vs Cobimetinib+Atezolizumab vs Regorafenib; Randomized II 363 Atezolizumab plus cobimetinib did not prolong overall survival (OS) vs. regorafenib NCT02788279 Bendell J, Ciardiello F, Tabernero J, et al. Annals of Oncology 2018; Volume 29, Issue suppl_5. mdy208.003, https://doi.org/10.1093/annonc/mdy208.003
Melanoma Atezolizumab+Cobimetinib+Vemur afenib vs Cobimetinib+Vemurafenib; Randomized III 513 Increase in Progression Free Survival and Duration of Response NCT02908672 N/A
Malignant Melanoma Sequencing schedule of Cobimetinib+Vemurafenib+Atezolizumab II 176 N/A NCT02902029 N/A
Multiple Myeloma Cobimetinib vs Cobimetinib+Venetoclax vs Cobimetinib+Venetoclax+Atezoliuzmab: Randomized I/II 72 N/A NCT03312530 N/A
Breast Cancer Atezolizumab+Cobimetinib vs Atezoluzimab+Idasanutlin; Randomized I/II 80 N/A NCT03566485 N/A
Advanced or Metastatic Solid Tumors Nivolumab vs Nivolumab+Ipilimumab vs Nivolumab+Ipilimumab+Cobimetinib (Pancreatic adenocarcinoma) I/II 162 N/A NCT01928394 N/A
Breast Cancer, Estrogen Receptor positive Breast Cancer Pre-operative window of opportunity study (Atezolizumab vs Atezolizumab+Cobimetinib vs Atezolizumab+Ipatasertib vs Atezolizumab+Ipatasertib+Bevacizumab) II 160 N/A NCT03395899 N/A
Selumetinib Advanced/Metastatic Solid tumors Selumetinib+Pembrolizumab dose escalation; non randomized I 50 N/A NCT03833427 N/A
Lung Cancer Intermittent selumetinib+durvalumab+Tremelimumab vs continuous selumetinib+Durvalumab+Tramelimumab; Randomized I/II 40 N/A NCT03581487 N/A
Advanced Solid Tumors Ascending doses Selumetinib+MEDI4736 vs Selumetinib+MEDI4736+Tremelimumab; Non-randomized I 58 N/A NCT02586987 N/A
Locally Advanced or Metastatic Non-Small-Cell Lung Cancer Gefitinib, AZD9291, or Selumetinib+Docetaxel + MEDI4736 vs Tremelimumab+MEDI4736; Randomized II 32 N/A NCT02179671 N/A
Binimetinib Colorectal Cancer Encorafenib, Binimetinib, Nivolumab; Single Group I/II 38 N/A NCT04044430 N/A
MSS, RAS-mutant Colorectal Cancer Binimetinib+Nivolumab vs Binimetinib+Nivolumab+Ipilimumab; Non-randomized Ib/II 90 N/A NCT03271047 N/A
Pancreatic Cancer, Non-Small Cell Lung Cancer Avelumab+Binimetinib vs Avelumab+Binimetinib+Talazoparib; Randomized II 127 N/A NCT03637491 N/A
Metastatic Colorectal Cancer Pembrolizumab+Binimetinib or Pembrolizumab+chemo vs Pembrolizumab+chemo+Binimetinib; Non-Randomized I 220 N/A NCT03374254 N/A
Breast Cancer Binimetinib+Avelumab vs anti-Ox40+Avelumab vs Utomilumab+Avelumab; Randomized II 150 N/A NCT03971409 N/A
Trametinib Colorectal Cancer, Malignant Neoplams of Digestive Organs Durvalumab+Trametinib; single group II 56 N/A NCT03428126 N/A
Non-Small Cell Lung Cancer, KRAS Gene Mutation Trametinib+Pembrolizumab vs Pembrolizumab+Trametinib; Non-Randomized I 42 N/A NCT03299088 N/A
Metastatic Melanoma Dabrafenib+Ipilimumab vs Dabrafenib+Trametinib+Ipilimumab; Randomized I 38 N/A NCT01767454 N/A
Melanoma ·Spartalizumab+Dabrafneib+Trametinib vs Dabrafenib+Trametinib; Randomized III 538 N/A NCT02967692 N/A
Melanoma Dose escalation; Durvalumab+Dabrafenib+Trametinib vs Durvalumab+Trametinib; Non-Randomized I 68 N/A NCT02027961 N/A
Melanoma BGB324+Pembrolizumab vs BGB324+Dabrafenib+Trametinib vs Pembrolizumab vs Dabrafenib+Trametinib; Randomized I/II 92 N/A NCT02872259 N/A
Melanoma Sequential Dabrafenib+Trametinib then Pembrolizumab vs Concurrent Dabrafenib+Trametnib and Pembrolizumab vs Pembrolizumab; Randomized II 60 N/A NCT02858921 N/A
Colorectal Cancer Nivolumab+Trametinib vs Nivolumab+Trametinib+Ipilimumab; Non-Randomized I/II 345 N/A NCT03377361 N/A
Melanoma Safety of Pembrolizumab+Trametinib+Dabrafenib; Randomized I/II 190 N/A NCT02130466 N/A
Metastatic Melanoma BRAF600 mutant Penbrolizumab+Dabrafenib+Trametinib; BRAFV600 WT Pembrolizumab+Trametinib; Non-Randomized II 50 N/A NCT03149029 N/A

In a phase 1 clinical trial across multiple tumor types, the combination of cobimetinib plus the PD-L1 inhibitor atezolizumab had manageable safety and demonstrated multiple objective responses in mismatch-repair proficient colorectal cancer (66). These results were particularly striking because MEK inhibitors and systemic immunotherapies have historically shown little or no activity in mismatch-repair proficient colorectal cancer as single agents. In this phase 1 study, there was no evident enrichment in of response in patients with KRAS/BRAF-mutant tumors versus wild-type tumors. These clinical findings provided support for the idea that MEK inhibitors are immunomodulatory and can sensitize immune excluded-tumors or immune-desert tumor types to respond to inhibitors of the PD-L1 pathway. Consistent with preclinical observations, paired pretreatment and cobimetinib on-treatment biopsies from this phase 1 study revealed an increase in CD8+ T-cell infiltration, suggesting successful translation of a preclinical finding into clinical development.

Based on these encouraging results, the combination of cobimetinib and atezolizumab were subsequently tested in the phase 3 colon cancer IMblaze370 study. In this study there was a trend towards longer overall survival for patients treated with cobimetinib plus atezolizumab over atezolizumab monotherapy, but the combination failed to meet its primary endpoint of superiority over a standard of care in this setting, regorafenib (13). Additionally, a phase III randomized study (IMspire170) investigating combination treatment with cobimetinib and atezolizumab (PD-L1 inhibitor) versus pembrolizumab (PD-1 inhibitor) in patients with BRAFV600 wild-type advanced melanoma failed to meet its primary endpoint of improved progression free survival with the combination (67). These two negative trials, one in a classically immune excluded-tumor tumor type (IMblaze370), and the other in a classically immune-inflamed tumor type (IMspire170), diminished initial hopes that this combination would be paradigm changing despite favorable effects of MEKi on the tumor immune microenvironment.

Other subsequent clinical trials of MEKi in combination with systemic immunotherapies did meet their primary outcomes, although none of these studies conclusively showed evidence of synergistic clinical activity. A smaller signal finding phase 2 trial in cholangiocarcinoma showed that the combination of cobimetinib plus atezolizumab significantly prolonged survival over atezolizumab monotherapy, but response rates in the combination treatment arm were low (68). In a separate phase 3 trial in BRAFV600E mutated melanoma (IMspire150) the combination of vemurafenib (B-Raf inhibitor), cobimetinib, and atezolizumab increased the median progression free survival from 10.6 months to 15.1 months when compared to treatment with vemurafenib, cobimetinib, and placebo (69). Intriguingly, overall response rates in the atezolizumab and control groups were very similar, and thus the progression free survival benefit of atezolizumab versus control was largely driven by the prolonged duration of response among the responders (21·0 vs 12·6 months). These results were similar to the phase 2 KEYNOTE-022 study, in which the combination of pembrolizumab plus dabrafenib and trametinib had longer median progression-free survival than placebo plus dabrafenib and trametinib. Together, these two studies indicate that checkpoint inhibition may deepen responses with targeted therapy in BRAFV600E melanoma. Although a late separation of overall survival curves favoring combination therapy was evident in both studies, the appropriate sequencing of immunotherapy and targeted therapy remains undetermined. Ongoing melanoma studies such as ImmunoCobiVem (NCT02902029), SECOMBIT (NCT02631447), DREAMseq (NCT02224781), COMBI-i (NCT02967692) may help to address clinical questions regarding the optimal sequence or combination of BRAF/MEK inhibition and immune checkpoint inhibitor therapy in BRAFV600E mutated melanoma. However, none of these studies will fully answer whether MEK inhibition and systemic immunotherapies have additive or synergistic activity.

Conclusions and Future Directions

Multiple preclinical studies have investigated the immunomodulatory effects of MEK inhibitors with, at times, contradictory conclusions (See Table 2). While there is agreement that MEKi can enhance the expression of MHC-I and antigen presentation, the mechanism(s) through which this occurs remains unclear. The effects of MEK inhibition on T-cell function is controversial and may be context-dependent, and the effects of MEKi on other immune populations remains largely unknown. The combination of MEKi with systemic immunotherapies shows potentially synergistic activity in preclinical models, but clinical studies have not demonstrated clear evidence that these agents are synergistic and MEKi has thus far failed in the clinic as a therapeutic strategy for transforming immune-resistant tumors into immune responsive tumors.

Table 2.

Summary of immunomodulatory effects, highlighting degree of confidence in the finding.

Category
of Change
Increased Decreased No Change
Finding Reference Type Of
Study
Finding Reference Type Of
Study
Finding Reference Type Of
Study
MHCI Expression Increased IFNg lead to increased MHC I expression Loi et al, 2015(19) In Vivo and In Vitro    
Brea et al, 2016(18) In Vitro
Angell et al, 2014(22) In Vitro
Dushyanthen et al, 2017(20) In Vivo and In Vitro
MEK inhibition leads to intrinsic upregulation of MHC I Brea et al, 2016(18) In Vitro
Angell et al, 2014(22) In Vitro
MEK inhibition leads to pAKT mediated MHC I upregulation Mimura et al, 2013(21) In Vitro
PDL1 Expression Increased PD-L1 Expression Loi et al, 2015(19) In Vivo and In Vitro Decreased PD-L1 Expression Jiang et al, 2013(70) In Vitro No Persistent Change in PD-L1 Expression Angell et al, 2014(22) In vitro
Cell Line Dependent Increased PD-L1 Expression Atefi et al, 2014(31) In Vitro Sumimoto et al, 2016(71) In Vitro Liu et al, 2015(30) In vitro
Cell Line Dependent Decreased PD-L1 Expression Atefi et al, 2014(31) In Vitro  
T Cell Infiltration Increased T Cell Homing Liu et at, 2015(30) In Vivo    
Ebert et al, 2016(45) In Vivo
T Cell Activation Increased CD4 T Cell Proliferation Liu et al, 2015(30) In Vivo Decreased T Cell Proliferation DeSilva et al, 1998(72) In Vitro In Vivo Cytokine Production Is Not Affected Hu-Lieskovan et al, 2015(44) In Vivo
Decreased exhaustion and apoptosis Ebert et al, 2016(45) In vitro and in vivo Dushyanthen et al, 2017(20) In Vivo and In Vitro No Affect On CD8+ T cells Ebert et al, 2016(45) In Vivo
Boni et al, 2010(39) In Vitro  
Vella et al, 2013(40) In Vitro
Allegrezza et al, 2016(41) In Vitro and In Vivo
Zwang et al. 2016(42) In Vitro
Decreased T Cell IFNg Production Boni et al, 2010(39) In Vitro
Dushyanthen et al, 2017(20) In Vivo and In Vitro
Hu-Lieskovan et al, 2015(44) In Vitro
Decreased Activation of Antigen-specific T cells Vella et al, 2013(40) In Vitro
Decreased T Cell IL2, TNFa, IL3 Production Dumont et al, 1998(43) In Vitro
Impaired tumor specific CD8+ T cell expansion and priming Ebert et al, 2016(45) In vivo
Tumor Cytokine Production Decreased Tumor IL10, IL6, VEGF Expression Sumimoto et al, 2006(34) In Vitro
Decreased Tumor Osteopotin Production Kim et al, 2014(35) In Vivo
Lin et al, 2015(36) In Vivo

More research is needed to understand why MEKi has broadly failed as an immunomodulatory strategy in the clinic despite success in multiple preclinical models. One possibility is that systemic MEKi has some potentially detrimental effects on anti-tumor immunity (eg a decrease PD-L1 expression thereby negating the benefit of αPD1/PD-L1 therapy, impaired T cell activation and priming), that can outweigh the positive effects of MEKi on the TME in some contexts. This highlights the need to further understand the therapeutic context(s) in which synergy is possible. Alternatively, MEKi plus PD-L1 may be insufficient to overcome the immunosuppressive phenotype of the tumors in which this strategy has been tested. More research is needed to determine mechanisms of resistance to MEKi plus αPD1/PD-L1 therapy, and to explore additional combinatorial strategies. Ongoing studies may further clarify the full effects of MEK inhibitors on immune cells, aiding in the design of more effective drug combinations that better harness the potential of MEK inhibitors.

Footnotes

Publisher's Disclaimer: This Author Accepted Manuscript is a PDF file of a an unedited peer-reviewed manuscript that has been accepted for publication but has not been copyedited or corrected. The official version of record that is published in the journal is kept up to date and so may therefore differ from this version.

References

Papers of particular interest, published recently, have been highlighted as:

• Of importance

•• Of major importance

  • 1.Dhillon AS, Hagan S, Rath O, Kolch W. MAP kinase signalling pathways in cancer. Oncogene. 2007. May 14;26(22):3279–90. [DOI] [PubMed] [Google Scholar]
  • 2.Liu F, Yang X, Geng M, Huang M. Targeting ERK, an Achilles’ Heel of the MAPK pathway, in cancer therapy. B. 2018;8(4):552–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Cargnello M, Roux PP. Activation and Function of the MAPKs and Their Substrates, the MAPK-Activated Protein Kinases. Microbiol Mol Biol Rev. 2011;75(1):50–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Hobbs GA, Der CJ, Rossman KL. RAS isoforms and mutations in cancer at a glance. J Cell Sci. 2016. April 1;129(7):1287–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Dong C, Davis RJ, Flavell RA. MAP KINASES IN THE IMMUNE RESPONSE. Annu Rev Immunol. 2002;20:55–72. [DOI] [PubMed] [Google Scholar]
  • 6.Yap JL, Worlikar S, MacKerell AD, Shapiro P, Fletcher S. Small-Molecule Inhibitors of the ERK Signaling Pathway: Towards Novel Anticancer Therapeutics. Vol. 6, ChemMedChem. 2011. p. 38–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Caunt CJ, Sale MJ, Smith PD, Cook SJ. MEK1 and MEK2 inhibitors and cancer therapy: the long and winding road. Nat Rev Cancer. 2015. October 1;15(10):577–92. [DOI] [PubMed] [Google Scholar]
  • 8.FDA approves encorafenib and binimetinib in combination for unresectable or metastatic melanoma with BRAF mutations ∣ FDA [Internet]. [cited 2020 Aug 27]. Available from: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-encorafenib-and-binimetinib-combination-unresectable-or-metastatic-melanoma-braf [Google Scholar]
  • 9.Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015;372(26):2509–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Kopetz S, Grothey A, Yaeger R, Van Cutsem E, Desai J, Yoshino T, et al. Encorafenib, Binimetinib, and Cetuximab in BRAF V600E–Mutated Colorectal Cancer. N Engl J Med. 2019. October 24;381(17):1632–43. [DOI] [PubMed] [Google Scholar]
  • 11.Larkin J, Ascierto PA, Dréno B, Atkinson V, Liszkay G, Maio M, et al. Combined Vemurafenib and Cobimetinib in BRAF -Mutated Melanoma. N Engl J Med. 2014. November 13;371(20):1867–76. [DOI] [PubMed] [Google Scholar]
  • 12.Wolchok JD, Chiarion-Sileni V, Gonzalez R, Rutkowski P, Grob J-J, Cowey CL, et al. Overall Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N Engl J Med. 2017;377(14):1345–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Eng C, Kim TW, Bendell J, Argilés G, Tebbutt NC, Di Bartolomeo M, et al. Atezolizumab with or without cobimetinib versus regorafenib in previously treated metastatic colorectal cancer (IMblaze370): a multicentre, open-label, phase 3, randomised, controlled trial. Lancet Oncol. 2019;20:849–61. [DOI] [PubMed] [Google Scholar]
  • 14.Agrawal S, Kishore MC. MHC class I gene expression and regulation. Vol. 9, Journal of Hematotherapy and Stem Cell Research. 2000. p. 795–812. [DOI] [PubMed] [Google Scholar]
  • 15.Yarchoan M, Johnson BA, Lutz ER, Laheru DA, Jaffee EM. Targeting neoantigens to augment antitumour immunity. Nat Rev Cancer. 2017. April 24;17(4):209–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Leffers N, Lambeck AJA, de Graeff P, Bijlsma AY, Daemen T, van der Zee AGJ, et al. Survival of ovarian cancer patients overexpressing the tumour antigen p53 is diminished in case of MHC class I down-regulation. Gynecol Oncol. 2008. September 1;110(3):365–73. [DOI] [PubMed] [Google Scholar]
  • 17.Watson NFS, Ramage JM, Madjd Z, Spendlove I, Ellis IO, Scholefield JH, et al. Immunosurveillance is active in colorectal cancer as downregulation but not complete loss of MHC class I expression correlates with a poor prognosis. Int J Cancer. 2006. January 1;118(1):6–10. [DOI] [PubMed] [Google Scholar]
  • 18.Brea EJ, Oh CY, Manchado E, Budhu S, Gejman RS, Mo G, et al. Kinase regulation of human MHC class i molecule expression on cancer cells. Cancer Immunol Res. 2016;4(11):936–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Loi S, Dushyanthen S, Beavis PA, Salgado R, Denkert C, Savas P, et al. RAS/MAPK activation is associated with reduced tumor-infiltrating lymphocytes in triple-negative breast cancer: Therapeutic cooperation between MEK and PD-1/PD-L1 immune checkpoint inhibitors. Clin Cancer Res. 2016;22(6):1499–509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Dushyanthen S, Teo ZL, Caramia F, Savas P, Mintoff CP, Virassamy B, et al. Agonist immunotherapy restores T cell function following MEK inhibition improving efficacy in breast cancer. Nat Commun. 2017;8(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Mimura K, Shiraishi K, Mueller A, Izawa S, Kua L-F, So J, et al. The MAPK pathway is a predominant regulator of HLA-A expression in esophageal and gastric cancer. J Immunol. 2013. December 15;191 (12):6261–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Angell TE, Lechner MG, Jang JK, LoPresti JS, Epstein AL. MHC class I loss is a frequent mechanism of immune escape in papillary thyroid cancer that is reversed by interferon and selumetinib treatment in vitro. Clin Cancer Res. 2014. December 1;20(23):6034–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Kono M, Dunn IS, Durda PJ, Butera D, Rose LB, Haggerty TJ, et al. Role of the mitogen-activated protein kinase signaling pathway in the regulation of human melanocytic antigen expression. Mol Cancer Res. 2006;4(10):779–92. [DOI] [PubMed] [Google Scholar]
  • 24.Goldberg MV, Maris CH, Hipkiss EL, Flies AS, Zhen L, Tuder RM, et al. Role of PD-1 and its ligand, B7-H1, in early fate decisions of CD8 T cells. Blood. 2007;110(1):186–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Freeman BGJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, et al. PD-1 Freeman JEM 2000. J Exp Med. 2000;192(7):1028–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012. April 1;12(4):252–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Wang Q, Liu F, Liu L. Prognostic significance of PD-L1 in solid tumor: An updated meta-analysis. Vol. 96, Medicine (United States). 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Coelho MA, de Carné Trécesson S, Rana S, Zecchin D, Moore C, Molina-Arcas M, et al. Oncogenic RAS Signaling Promotes Tumor Immunoresistance by Stabilizing PD-L1 mRNA. Immunity. 2017;47(6):1083–1099.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Fey D, Croucher DR, Kolch W, Kholodenko BN. Crosstalk and signaling switches in mitogen-activated protein kinase cascades. Front Physiol. 2012;3:355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Liu L, Mayes PA, Eastman S, Shi H, Yadavilli S, Zhang T, et al. The BRAF and MEK inhibitors dabrafenib and trametinib: Effects on immune function and in combination with immunomodulatory antibodies targeting PD-1, PD-L1, and CTLA-4. Clin Cancer Res. 2015;21(7):1639–51. [DOI] [PubMed] [Google Scholar]
  • 31.Atefi M, Avramis E, Lassen A, Wong DJL, Robert L, Foulad D, et al. Effects of MAPK and PI3K pathways on PD-L1 expression in melanoma. Clin Cancer Res. 2014;20(13):3446–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Liu C-Z, Zhang L, Chang X-H, Cheng Y-X, Cheng H-Y, Ye X, et al. Overexpression and Immunosuppressive Functions of Transforming Growth Factor 1, Vascular Endothelial Growth Factor and Interleukin-10 in Epithelial Ovarian Cancer. Chin J Cancer Res. 2012;24(2):130–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Koido S, Homma S, Hara E, Namiki Y, Takahara A, Komita H, et al. Regulation of tumor immunity by tumor/dendritic cell fusions. Vol. 2010, Clinical and Developmental Immunology. Hindawi Publishing Corporation; 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.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. July 10;203(7):1651–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kim EK, Jeon I, Seo H, Park YJ, Song B, Lee KA, et al. Tumor-derived osteopontin suppresses antitumor immunity by promoting extramedullary myelopoiesis. Cancer Res. 2014;74(22):6705–16. [DOI] [PubMed] [Google Scholar]
  • 36.Lin CN, Wang CJ, Chao YJ, Lai MD, Shan YS. The significance of the co-existence of osteopontin and tumor-associated macrophages in gastric cancer progression. BMC Cancer. 2015;15(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Allegrezza MJ, Rutkowski MR, Stephen TL, Svoronos N, Perales-Puchalt A, Nguyen JM, et al. Trametinib drives T-cell-dependent control of KRAS-mutated tumors by inhibiting pathological myelopoiesis. Cancer Res. 2016;76(21):6253–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Smith-Garvin JE, Koretzky GA, Jordan MS. T cell activation. Vol. 27, Annual Review of Immunology. 2009. p. 591–619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Boni A, Cogdill AP, Dang P, Udayakumar D, Njauw CNJ, Sloss CM, et al. Selective BRAFV600E inhibition enhances T-cell recognition of melanoma without affecting lymphocyte function. Cancer Res. 2010;70(13):5213–9. [DOI] [PubMed] [Google Scholar]
  • 40.Vella LJ, Pasam A, Dimopoulos N, Andrews M, Puaux A-L, Louahed J, et al. MEK inhibition, alone or in combination with BRAF inhibition, impairs multiple functions of isolated normal human lymphocytes and dendritic cells. J Immunother Cancer. 2013;1 (S1). [DOI] [PubMed] [Google Scholar]
  • 41.Allegrezza MJ, Rutkowski MR, Stephen TL, Svoronos N, Tesone AJ, Perales-Puchalt A, et al. IL-15 AGONISTS OVERCOME THE IMMUNOSUPPRESSIVE EFFECTS OF MEK INHIBITORS HHS Public Access. Cancer Res. 2016;76(9):2561–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Zwang NA, Zhang R, Germana S, Fan MY, Hastings WD, Cao A, et al. Selective Sparing of Human Tregs by Pharmacologic Inhibitors of the Phosphatidylinositol 3-Kinase and MEK Pathways. Am J Transplant. 2016;16(9):2624–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Dumont FJ, Staruch MJ, Fischer P, DaSilva C, Camacho R. Inhibition of T cell activation by pharmacologic disruption of the MEK1/ERK MAP kinase or calcineurin signaling pathways results in differential modulation of cytokine production. J Immunol. 1998;160(6):2579–89. [PubMed] [Google Scholar]
  • 44.Hu-Lieskovan S, Mok S, Homet Moreno B, Tsoi J, Robert L, Goedert L, et al. Improved antitumor activity of immunotherapy with BRAF and MEK inhibitors in BRAFV600E melanoma. Sci Transl Med. 2015;7(279). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Ebert PJR, Cheung J, Yang Y, McNamara E, Hong R, Moskalenko M, et al. MAP Kinase Inhibition Promotes T Cell and Anti-tumor Activity in Combination with PD-L1 Checkpoint Blockade. Immunity. 2016;44(3):609–21. [DOI] [PubMed] [Google Scholar]
  • 46.Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Vol. 12, Nature Reviews Immunology. 2012. p. 253–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Ugel S, De Sanctis F, Mandruzzato S, Bronte V. Tumor-induced myeloid deviation: when myeloid-derived suppressor cells meet tumor-associated macrophages. J Clin Invest. 2015. September;125(9):3365–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Steinberg SM, Shabaneh TB, Zhang P, Martyanov V, Li Z, Malik BT, et al. Myeloid cells that impair immunotherapy are restored in melanomas which acquire resistance to BRAF inhibitors. Cancer Res. 2017;77(7):1599–610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Poon E, Mullins S, Watkins A, Williams GS, Koopmann JO, Di Genova G, et al. The MEK inhibitor selumetinib complements CTLA-4 blockade by reprogramming the tumor immune microenvironment. J Immunother Cancer. 2017;5(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Sarvaria A, Madrigal A, Saudemont A. REVIEW B cell regulation in cancer and anti-tumor immunity. Cell Mol Immunol. 2017;14:662–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Lebien TW, Tedder TF. B lymphocytes: how they develop and function. Blood. 2008. September 1;112(5):1570–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Yanaba K, Bouaziz JD, Haas KM, Poe JC, Fujimoto M, Tedder TF. A Regulatory B Cell Subset with a Unique CD1dhiCD5+ Phenotype Controls T Cell-Dependent Inflammatory Responses. Immunity. 2008;28(5):639–50. [DOI] [PubMed] [Google Scholar]
  • 53.Rosser EC, Mauri C. Regulatory B Cells: Origin, Phenotype, and Function. Vol. 42, Immunity. 2015. p. 607–12. [DOI] [PubMed] [Google Scholar]
  • 54.Yarchoan M, Mohan AA, Dennison L, Vithayathil T, Ruggieri A, Lesinski GB, et al. MEK inhibition suppresses B regulatory cells and augments anti-tumor immunity. PLoS One. 2019;14(10). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Sharma P, Allison JP. The future of immune checkpoint therapy. Science. 2015. April 3;348(6230):56–61. [DOI] [PubMed] [Google Scholar]
  • 56.Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: A common denominator approach to cancer therapy. Vol. 27, Cancer Cell. 2015. p. 450–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Kyi C, Postow MA. Checkpoint blocking antibodies in cancer immunotherapy. FEBS Lett. 2014. January 21;588(2):368–76. [DOI] [PubMed] [Google Scholar]
  • 58.Melero I, Berman DM, Aznar MA, Korman AJ, Gracia JLP, Haanen J. Evolving synergistic combinations of targeted immunotherapies to combat cancer. Nat Rev Cancer. 2015. August 1;15(8):457–72. [DOI] [PubMed] [Google Scholar]
  • 59.Salazar-Fontana LI, Bierer BE. T-lymphocyte coactivator molecules. Curr Opin Hematol. 2001;8(1):5–11. [DOI] [PubMed] [Google Scholar]
  • 60.Croft M Co-stimulatory members of the TNFR family: keys to effective T-cell immunity? Nat Rev Immunol. 2003. August 1;3(8):609–20. [DOI] [PubMed] [Google Scholar]
  • 61.Redmond WL, Ruby CE, Weinberg AD. Role of OX40 in T cell costim and survival. Crit Rev Immunol. 2009;29(3):187–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Melero I, Shuford WW, Newby SA, Aruffo A, Ledbetter JA, Hellström KE, et al. Monoclonal antibodies against the 4-1BB T-cell activation molecule eradicate established tumors. Nat Med. 1997. June 1;3(6):682–5. [DOI] [PubMed] [Google Scholar]
  • 63.Shuford WW, Klussman K, Tritchler DD, Loo DT, Chalupny J, Siadak AW, et al. 4-1BB costimulatory signals preferentially induce CD8+ T cell proliferation and lead to the amplification in vivo of cytotoxic T cell responses. J Exp Med. 1997. July 7;186(1):47–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.So T, Choi H, Croft M. OX40 Complexes with Phosphoinositide 3-Kinase and Protein Kinase B (PKB) To Augment TCR-Dependent PKB Signaling. J Immunol. 2011;186(6):3547–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Ventola CL. Cancer Immunotherapy, Part 3: Challenges and Future Trends. P T. 2017. August;42(8):514–21. [PMC free article] [PubMed] [Google Scholar]
  • 66.Hellmann MD, Kim T, Lee CB, Goh B, Miller WH Jr, Oh D, et al. Phase Ib study of atezolizumab combined with cobimetinib in patients with solid tumors. Prod Dev Oncol. 2019;14:1134–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Arance AM, Gogas H, Dreno B, Flaherty KT, Demidov L, Stroyakovskiy D, et al. Combination treatment with cobimetinib (C) and atezolizumab (A) vs pembrolizumab (P) in previously untreated patients (pts) with BRAFV600 wild type (wt) advanced melanoma: Primary analysis from the phase III IMspire170 trial. Ann Oncol. 2019;30:v906. [Google Scholar]
  • 68.Yarchoan M, Cope L, Anders RA, Noonan A, Goff L, Goyal L, et al. A multicenter randomized phase 2 trial of atezolizumab as monotherapy or in combination with cobimetinib in biliary tract cancers (BTCs): A NCI Experimental Therapeutics Clinical Trials Network (ETCTN) study [Internet]. AACR. 2020. [cited 2020 Aug 12]. Available from: https://www.abstractsonline.com/pp8/#!/9045/presentation/10752 [Google Scholar]
  • 69.Gutzmer R, Stroyakovskiy D, Gogas H, Robert C, Lewis K, Protsenko S, 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(10240):1835–44. [DOI] [PubMed] [Google Scholar]
  • 70.Jiang X, Zhou J, Giobbie-Hurder A, Wargo J, Hodi FS. The activation of MAPK in melanoma cells resistant to BRAF inhibition promotes PD-L1 expression that is reversible by MEK and PI3K inhibition. Clin Cancer Res. 2013. February 1;19(3):598–609. [DOI] [PubMed] [Google Scholar]
  • 71.Sumimoto H, Takano A, Teramoto K, Daigo Y. RAS-mitogen-activated protein kinase signal is required for enhanced PD-L1 expression in human lung cancers. LoS One. 2016;11(11). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.DeSilva DR, Jones EA, Favata MF, Jaffee BD, Magolda RL, Trzaskos JM, et al. Inhibition of mitogen-activated protein kinase kinase blocks T cell proliferation but does not induce or prevent anergy. J Immunol. 1998;160(9):4175–81. [PubMed] [Google Scholar]

RESOURCES