Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Feb 1.
Published in final edited form as: Hematol Oncol Clin North Am. 2020 Oct 26;35(1):111–128. doi: 10.1016/j.hoc.2020.09.005

Advanced Melanoma: Resistance Mechanisms to Current Therapies

Douglas B Johnson 1, April KS Salama 2, Alexandra M Haugh 3
PMCID: PMC7991196  NIHMSID: NIHMS1641335  PMID: 33759769

Summary

Despite the recent advances in melanoma treatment, resistance to currently available immune and targeted therapies remains the major barriers to long-term disease control in many patients. Most often, this is associated with intrinsic/innate resistance to immune checkpoint inhibitors and acquired resistance to BRAF and MEK kinase inhibitors. Mechanisms of resistance are diverse and will require innovative and perhaps personalized management strategies.

Keywords: immune checkpoint inhibitor, immunotherapy, resistance, BRAF inhibitor, MEK inhibitor, melanoma

Introduction

Advances over the last decade have brought about remarkable changes in the treatment of metastatic melanoma. The introduction of targeted therapies including BRAF and MEK inhibitors as well as immune checkpoint inhibitors (ICI) led to dramatically improved outcomes compared with chemotherapy, which had not been shown to improve survival in any large randomized trials. While impressive outcomes are seen in some patients, responses to these agents are heterogeneous. Initial robust responses to BRAF/MEK inhibitors are complicated by eventual disease progression in most treated patients. Immune checkpoint inhibitors may induce more durable long-term responses in some cases, yet a significant proportion of patients do not respond to these agents (innate resistance) or respond transiently followed by progression (acquired resistance). This review provides an overview of recent insights into innate and acquired resistance to both targeted therapies and checkpoint inhibition in metastatic melanoma. Further understanding of the mechanisms involved in resistance is warranted to inform optimal clinical decision making in individual patients as well as to drive therapeutic advances that continue to improve outcomes.

Discussion

Resistance to Checkpoint Inhibitors

Mutational and Neo-antigen Burden:

Several studies have demonstrated a correlation between higher tumor mutational burden and response to checkpoint inhibition with either anti-PD-1/PD-L1 or anti-CTLA-4.17 Malignant cells are often detected by immune surveillance through recognition of tumor-derived neoantigens that are foreign and thus immunogenic.8 As these neoantigens are a result of somatic mutations, higher mutational load is thought to contribute to a wider and more robust spectrum of neoantigens that can be recognized by the immune system. Tumors deficient in mismatch repair, regardless of tissue of origin, are also particularly sensitive to immune checkpoint inhibition.911 These tumors are characterized by a very high frequency of somatic alterations, with a mutational load 10 to 100-fold greater than those that are mismatch-proficient, which likely plays a role in response to ICI.12

Recognition of a specific neoantigen may be complicated by the spectrum of different mutations within clusters of malignant cells in each tumor. Clonal neoantigen burden has been associated with response to ICI while a high burden of sub-clonal alterations was seen in tumors that progressed on therapy.13,14 Cytotoxic chemotherapy may potentiate production of sub-clonal neoantigens, possibly contributing to decreased efficacy of ICI in some patients despite an overall high mutational burden.14 Neoantigens that arise earlier in tumorigenesis and are shared by a majority of cancer cells are likely to trigger a more robust immune response.

Most melanoma tumors are driven by UV-induced mutagenesis with a high frequency of somatic mutations. Subtypes of melanoma that are less likely to be UV-mediated, such as mucosal or acral melanoma, tend to have fewer point mutations but more frequent somatic structural or copy number aberrations.15 Given the lower overall mutational burden and likely decreased neoantigen production, it is speculated that these subtypes may be less likely to respond to immune checkpoint inhibitors. Indeed, several studies report lower response rates in these non-cutaneous melanomas.1618 Conversely, desmoplastic melanomas, which have the highest mutational burdens of all melanomas, also have the highest response rates.19 Miao et al stratified melanoma tumors based on mutational signature, which included UV-associated, those associated with exposure to alkylating agents and tumors not clearly associated with specific environmental exposures. When stratified based on dominant mutational signature, there was no significant difference in mutational burden between patients with progressive disease and those who responded to immunotherapy.13 The authors thus postulated that mutational burden may actually serve as a marker of underlying pathobiology that promotes immunogenicity rather than a true mechanism of response to therapy in melanoma. In addition, most studies have suggested threshold effects rather than a linear relationship between response and TMB within individual tumor types.

Tumors can demonstrate selective loss of neoantigens recognized by T-cells as a mechanism to avoid immune detection.20,21 Malignant cells that express these neoantigens are selectively lost from the overall tumor population through the loss of mutant alleles or epigenetic silencing of implicated genes.20 Neoantigens lost by tumor cells following ICI were noted to have higher affinity for MHC variants and result in stronger TCR responses in peripheral lymphocytes than those that were retained.22 The loss of T-cell recognized neoantigens correlates with the development of neo-antigen specific reactivity amongst tumor infiltrating lymphocytes, suggesting that T-cells likely play a role in modulating immunoediting.20

PD-L1 Expression

PD-L1 expression in tumor cells or tumor-infiltrating immune cells has unsurprisingly been correlated with response to PD-1/PD-L1 blockade in melanoma.2326 PD-L1 negativity, however, is not a definitive marker of resistance and studies have consistently demonstrated durable responses in some patients with PD-L1 negative tumors.24,25,27 PD-L1 status is used to identify patients appropriate for treatment with immunotherapy in some malignancies but is of uncertain utility in melanoma, which is also associated with relatively high rates of PD-L1 positivity.28 PD-L1 expression is frequently heterogenous over time, within individual patients or even within the same tumor. Further, several different IHC stains and cut-off values were employed in early trials, further complicating assessment of PD-L1 status.28 Expression of PD-1 and PD-L1 are also known to be dynamic and influenced by several complex and inter-related factors within the tumor microenvironment, including IFN or other cytokine signaling, genomic alterations or changes induced by radiation or targeted therapy.12,29

Specific Genomic Alterations

Tumors that are subject to immune surveillance may become enriched for a wide range of genomic alterations that aid in immune escape. Alterations in the gene encoding β−2-microglobulin (B2M), the invariant chain of MHC, have been shown to play a role in both innate and acquired resistance to checkpoint inhibition.3032 Decreased expression of MHC, whether brought about by genomic changes or other mechanisms, has been associated with lack of response to anti-PD1 (MHC-II) and anti-CTLA-4 (MHC-1).33 Germline differences in HLA genotype may impact response to checkpoint inhibition, potentially due to a decreased spectrum of neoantigens presented by tumors with homozygous HLA alleles.34

Loss of function mutations in JAK1/JAK2 have been associated with both innate and acquired resistance to PD-1/PD-L1 inhibitors in melanoma.32,35 Tumors with JAK1/JAK2 mutations demonstrate a lack of response to IFN-γ stimulation, including a lack of associated PD-L1 expression.32,35 Several genomic defects in the IFN-γ pathway have also been identified amongst tumors that do not respond to CTLA-4 blockade.36 Overall these mutations appear uncommon, and likely only explain a minor proportion of resistance.

Certain tumor signaling pathways have also been shown to suppress the recruitment and diversification of T cells within the tumor microenvironment.37,38 Activation of the WNT/β-catenin pathway in melanoma contributed to T cell exclusion in mouse models, evidenced by a complete lack of T-cell infiltrate in many tumors.37 Single cell RNA sampling of melanoma tumor cells treated with ICI identified a specific transcription factor, TCF7, that served as a marker of response regardless of the extent of lymphocytic invasion. This transcription factor plays a role in the Wnt/β-catenin signaling pathway and is involved with cytotoxic T-cell auto-renewal, differentiation and persistence.39 The loss of PTEN within tumor cells has been correlated with decreased T-cell infiltration, increased expression of immunosuppressive cytokines and overall inferior clinical response to ICI.40,41

Recent evidence has suggested that the MAPK pathway may play a role in immune escape through upregulation of VEGF and other immunosuppressive cytokines in addition to other unknown mechanisms.42,43 BRAF inhibitors have been shown to increase intra-tumoral T-cell infiltration and result in a more favorable TME with fewer immunosuppressive cytokines, fewer myeloid-derived suppressor stem cells and decreased PD-L1 expression, while other studies have suggested a low CD8+ T cell infiltrate at the time of progression on BRAF inhibitors.4448 Acquired resistance to BRAF inhibition has been associated with upregulation of PD-L1 on tumor cells and treatment with MAPK inhibitors can induce transcriptional signatures similar to the IPRES signature associated with ICI resistance.47,49 It remains unclear if and how changes induced by BRAF inhibition impact subsequent response to ICI in melanoma yet molecular evidence is suggestive of overlapping resistance mechanisms to both classes of therapy.50,51 Retrospective analysis found higher response rates to first-line immunotherapy amongst NRAS-mutant melanoma compared with BRAF or BRAF/NRAS wild-type melanoma although these data have not been validated extensively.52 The impact of MAPK pathway alterations in the development of ICI resistance is yet to be defined.

Tumor Microenvironment

Malignant cells employ a variety of mechanisms to evade immune destruction, many of which involve alterations in the surrounding tumor microenvironment that create an immunosuppressive barrier. The presence of tumor infiltrating lymphocytes within the TME is associated with improved outcomes across wide spectrum of malignancies.53 Higher numbers of intra-tumoral CD8+ T cells, both in the core and at the invasive margin, are predictive of response to anti-PD-1 therapy in melanoma, particularly if they express PD-1.27,54 Functional analysis of TILs that expressed high amounts of both PD-1 and CTLA-4 revealed a partially exhausted CD8+ T-cell phenotype that could be restored to a fully activated state upon PD-1/PD-L1 inhibition.27

Persistent antigen exposure and activation is complicated by eventual T-cell exhaustion both in the setting of ICI therapy and natural immune surveillance. The interaction between PD-1 and PD-L1 contributes significantly to the loss of effector T-cell function exhibited by exhausted CD8+ T-cells.55 Blockade of PD-L1/PD-1 can re-invigorate exhausted cytotoxic T-cells; an abundance of partially exhausted CD8+ T-cells within the TME has been shown to correlate with response to ICI.27

These re-invigorated T-cells, however, will revert back to an exhausted state in the face of continued and persistent antigen exposure and do not exhibit a memory T-cell phenotype upon antigen clearance.56 Recent studies have uncovered a hardwired epigenetic profile unique to exhausted T-cells that may limit prolonged response to checkpoint inhibition and contribute to acquired resistance.57,58 Epigenetic therapies have been proposed as a potential mechanism to overcome T-cell exhaustion and are being evaluated as an adjunct to ICI in refractory patients in ongoing trials.

The PD-1/PD-L1 interaction drives adaptive resistance through several additional mechanisms, many of which are the result of negative feedback interactions within the TME that upregulate PD-L1 expression.12 PD-L1 plays a role in induction and maintenance of regulatory T-cells and immunosuppressive myeloid cells within the TME, which are of uncertain significance in predicting response to ICI.59,60 CTLA-4 inhibitors have been shown to result in upregulation of CD4(+)Foxp3(−) T cells within the tumor microenvironment, which express PD-1 but lack cytotoxic function and express Treg-associated markers.61 Treatment with both PD-1/PD-L1 and CTLA-4 antibodies notably mitigates this effect in many cases and persistence of these cells after anti-PD-1 correlates with poor prognosis.62

A group of 26 transcriptomic signatures, collectively referred to as the innate anti-PD-1 resistance (IPRES) signature, were found to be co-enriched in pre-treatment ICI-resistant melanomas.63 This signature donates upregulation of genes involved with mesenchymal transition, angiogenesis and wound healing. Epithelial to mesenchymal transition has been associated with an increased presence of various immune checkpoint molecules, regulatory T cells and immunosuppressive cytokines.6466 The epithelial to mesenchymal transition signature is notably associated with TNFα, which promotes phenotypic plasticity and upregulation of an innate resistance signature through translational reprogramming.67

Both type I and II interferon signaling play an important role in T-cell activation required for initial response to checkpoint inhibition yet prolonged IFN signaling has also been associated with immune supression.68 Interferon signaling accordingly plays a complicated role in modulating response to checkpoint inhibition. Increased expression of IFN-γ and IFN-related genes has been associated with response to anti-PD-1 in melanoma and defects in IFN signaling pathways have been associated with acquired resistance.32,69 Gene expression studies have also identified an interferon-γ related GEP that was necessary, but not always sufficient, for clinical benefit across tumor types exposed to ICI.70

Persistent IFN signaling induces epigenomic and genomic alterations within tumors that dampen immune response, including expression of PD-L1 as well as upregulation of multiple additional T-cell inhibitory receptors.71 IFN-γ may also play a role in expediting cytotoxic T-cell dependent immunoediting.72 As noted above, response to anti-PD-1 has been associated with a T-cell rich inflammatory TME, characterized by an abundance of TILs and PD-L1 expression. A responsive TME is also likely one that is “adaptive immune resistant” and further categorization of the cellular and molecular changes associated with adaptive immunity may help inform treatment decisions or uncover new therapeutic targets.70

Proteomic profiling found immunotherapy-responsive tumors to be enriched for proteins involved with oxidative phosphorylation and lipid metabolism compared with non-responsive samples. These samples were also enriched for antigen presentation and IFN signaling. Functional studies demonstrated that increased lipid metabolism led to upregulation of antigen expression by melanoma cells, increasing immunogenicity and potential for ICI response.73

The Host Microbiome

Increasing evidence suggests that the intestinal microbiome plays an important role in a wide variety of inflammatory or immune-mediated conditions. Increased microbial biodiversity within the host microbiome has been associated with response to PD-1 inhibition in melanoma.74 Specific bacterial species have also been noted to be prevalent amongst responders while others are associated with a lack of response.7476 Xenografted germ-free mice transplanted with stool samples from responding patients demonstrated improved outcomes with anti-PD-1 compared with those transplanted from non-responders.76 Concurrent and prior antibiotic therapy has also been correlated with decreased response to ICI.77,78 High dietary fiber intake has been shown to contribute to an immunostimulatory landscape within the microbiome and patients with melanoma who self-reported a high fiber diet were significantly more likely to respond to anti-PD-1 agents.79,80

Overcoming ICI Resistance

Several strategies have been proposed to augment cytotoxic T-cell priming and tumor infiltration integral to ICI response, which were in part mentioned above. The full spectrum of current combination therapy approaches are beyond the scope of this review. Radiation therapy broadens the spectrum of T-cell-receptors amongst tumor-infiltrating lymphocytes, which may help to overcome innate or acquired mechanisms of resistance involving T-cell exclusion.81 Melanoma vaccines may also augment the effect of checkpoint inhibition, particularly when directed towards particular neoantigens associated with a robust response to ICI.7,82 The combination of checkpoint inhibitors and cancer vaccines, including individualized vaccines directed at neoantigens shown to demonstrate immunogenicity within a specific tumor, are currently under investigation in early phase trials.82

Certain oncolytic viruses capitalize on defective interferon signaling to enter and replicate within cells and may therefore be used to target IFN- γ deficient clones and overcome associated resistance.83 Agonists of the stimulator for interferon genes (STING) receptor may increase sensitivity to checkpoint inhibition by increasing PD-L1 expression through upregulation of the JAK/STAT pathway.84,85 A resistance program associated with T cell exclusion and immune evasion identified by single cell RNA sequencing was also found to be repressed by CDK4/6 inhibition when given in combination with ICI in mouse models.86 Other approaches, including co-stimulation of additional immune checkpoints, toll-like receptor agonists, and tumor infiltrating lymphocytes may also have promise. Antibodies targeting the T-cell inhibitory receptor lymphocyte activation gene-3 (LAG-3) showed promise when used with nivolumab in melanoma refractory to anti-PD-1/PD-L1 and are being evaluated with anti-CTLA-4 in this setting as well as with anti-PD-1/PD-L1 in ICI-naïve patients (NCT03978611, NCT03743766).87 Engagement of toll-like receptors within the TME promotes innate immune activation and associated pro-inflammatory cytokine production that can increase intra-tumoral T-cell infiltration and potentiate response to ICI.88 Early phase trials have shown encouraging results in anti-PD-1/PDL-1 refractory patients treated with an intra-tumoral TLR9 agonist and ipilimumab with an overall response rate of 47% (7 of 15 patients).89 Adoptive cell therapy with autologous tumor-infiltrating lymphocytes has been attempted and refined in melanoma for several decades with more recent trials limited to those who have progressed on standard therapies. TIL therapy optimized with pre-infusion lymphodepletion and a post-transfer IL-2 regimen resulted in durable response in some refractory patients with an ORR of 36.4% and a median duration of response that was not reached after 17 months.90

Resistance to BRAF/MEK Inhibitors

Mutations in BRAF occur in almost half of all melanomas and thus represent a major therapeutic target. A majority of BRAF-mutant melanomas harbor a substitution at codon 600 of the BRAF gene, which results in constitutive kinase activation and downstream activation of the MAPK pathway. The MAPK pathway plays a role in a wide spectrum of intracellular processes, including differentiation, stress response, and cell survival, which it also regulates via physiologic negative feedback mechanisms. Constitutive activation of the MAPK pathway results in unregulated cell growth and proliferation that drives tumorigenesis.

The development of small molecule inhibitors specific to the BRAF-mutant kinase (BRAFi) represented a major breakthrough in the treatment of melanoma. These agents were associated with significantly improved overall response rates, PFS and OS when compared with chemotherapy in patients with BRAF V600 mutant melanoma.91,92 Primary resistance to BRAF inhibition is relatively rare, with impressive initial response in many patients, yet these regimens are frequently complicated by acquired resistance. Unlike many other malignancies treated with other targeted kinase inhibitors, which often develop resistance through “gate-keeper” mutations that impede interaction between the inhibitor and the mutated kinase, BRAF-mutated melanomas employ several complex alternative mechanisms to upregulate the downstream MAPK pathway and bring about resistance.

Evidence that re-activation of the MAPK pathway was involved with acquired resistance to BRAFi led to the introduction of combined therapy with both BRAF and MEK inhibition (MEKi). MEK is directly down-stream from BRAF in the MAPK cascade and MEKi was independently associated with superior outcomes compared with chemotherapy in patients with BRAF V600 mutant melanoma.93 Targeting multiple sites along the MAPK pathway with the addition of MEKi was proposed in an attempt to mitigate eventual resistance as well as to promote a more robust response to therapy. Multiple trials demonstrated significant improvement in response rate, PFS and OS with both BRAF and MEK inhibitors compared with BRAFi monotherapy and combination therapy became standard of care in BRAF V600 mutant melanoma.9496

Despite significant improvements with combination therapy, BRAF/MEK inhibition is still frequently complicated by eventual resistance, most often via upregulation of MAPK signaling. Although many studies have focused on categorizing BRAF inhibitor resistance in melanoma, resistance to the BRAF/MEKi combination is due to similar mechanisms.9799 Eventual resistance to BRAF/MEKi should be expected in most patients, yet recently published follow-up data from the COMBI-v and COMBI-d trials suggests the potential for long-term benefit in a minority of patients treated with BRAF + MEK inhibitiors.100 Patients who demonstrated a complete response to therapy (109 of 563, 19%) had a 5-year overall survival of 71% compared with 34% in the overall cohort. Several baseline factors were also found to be associated with prolonged progression-free survival including older age, female sex, normal lactate dehydrogenase level, and less than three organ sites with distant metastasis. Patients who demonstrated a complete response shared similar baseline factors. PFS at 5 years was observed in approximately 15% of the population.100 Longer-term follow-up of patients with sustained response to BRAF/MEKi as well as further molecular and clinical characterization of this sub-group is warranted.

The categorization of BRAF/MEKi resistance is complicated by a diverse array of mechanisms with significant heterogeneity between patients and within individual tumors. While a single predominant resistance mechanism may be identified in one resistant tumor sample, additional biopsies from the same patient often demonstrate distinct or unknown drivers of resistance.99 Melanoma clones emerging after BRAFi therapy demonstrate branched evolution and some tumors can proliferate in the setting of BRAFi in the absence of any clear genomic driver.101 Intra-tumor heterogeneity can be explained in part by a suspected multi-step pattern of resistance acquisition. This starts with adaptive transcriptional reprogramming that allows for cell survival in the presence of BRAFi via phenotypic plasticity as well as increased signaling in alternative RTK pathways that frequently converge with the MAPK pathway. This adaptive transcriptional state allows tumor cells to survive long enough to acquire “fixed” mediators of resistance, which are often genomically mediated.

Increasing evidence suggests melanoma cells exposed to BRAF inhibition may capitalize on an innate stress reaction that promotes transition to a “slow-growth” phenotypic state associated with oncogene-induced senescence and de-differentiation as well as changes in chromatin remodeling and histone deacetylase activity.102104 Transcriptomic analysis of BRAF/MEKi resistant tumors demonstrated recurrent involvement of specific genes and pathways, which frequently demonstrated differential methylation of tumor cell-intrinsic CpG sites.105

The early adaptive “persister” state adopted by some melanoma cells in response to BRAF inhibition has also been associated with phenotypic transition to a more de-differentiated mesenchymal state.106,107 Overexpression of the transcription factor c-JUN has been associated with a mesenchymal gene signature and an EMT-like phenotypic transition signature in melanoma cells.108,109 Inhibition of c-JUN, either via direct silencing or upstream inhibition of c-JUN amino-terminal kinase (JNK), has been shown to increase overall cell death and decrease the population of “persister” cells when used in combination with BRAF inhibitors.106,108,109 Microphthalmia associated transcription factor (MITF), a transcription factor that controls multiple genes integral to melanocyte function, has been repeatedly implicated in BRAF/MEK inhibitor resistance. While some studies have implicated MITF up-regulation as a mechanism of BRAF resistance, a majority have demonstrated the emergence of “MITF-low” populations early in the course of acquired resistance.110113

In addition to re-activation of MAPK signaling, increased signaling in the PI3 kinase/AKT pathway has also been implicated in de novo and acquired resistance to BRAF/MEK inhibition in almost 20% of melanoma patients.114 MAPK pathway inhibition has been shown to induce upregulation of AKT signaling in resistant cells and high levels of AKT activity have been correlated with a lack of response to MEK inhibition in patients with BRAF-mutant melanoma.114116 Despite encouraging pre-clinical data, inhibitors of PI3K or downstream PI3K pathway effector molecules have largely failed to offer additional clinical benefit when used in combination with BRAF/MEK inhibitors in early stage trials.117

Increased signaling in the MAPK and PI3K pathways in resistant cells is modulated by multiple additional RTK pathways that are frequently up-regulated in the setting of BRAF/MEKi resistance.118123 Evidence suggests that treatment with BRAFi more likely results in the coordinated upregulation of multiple RTKs in individual tumor cells rather than selective upregulation of specific receptors that may be amenable to therapeutic intervention.117 Additional RTK pathways shown to be upregulated in resistant cells include epidermal growth factor receptor (EGFR), ERBB3, hepatocyte growth factor receptor (c-MET), platelet derived growth factor (PDGFR)-a, and the insulin like growth factor (IGF)-1 receptor.118123 Some studies have demonstrated an inverse correlation between MITF expression in resistant samples and upregulation of multiple RTKs, including the RTK AXL, which is over-expressed in many advanced malignancies and often associated with acquired resistance to chemotherapy.110112,124

Melanoma cells capable of adapting to MAPK inhibition will eventually acquire permanent genomic alterations that confer resistance to therapy. Genomic profiling studies have demonstrated a wide spectrum of genetic drivers associated with MAPKi resistance with significant intra-patient and intra-tumoral heterogeneity. As expected, a majority of identified mutations are associated with increased signaling within the MAPK pathway, which is restored in an estimated 80% of patients resistant to dabrafenib or vemurafenib.125 These include activating mutations in NRAS and/or MEK1/2 as well as BRAF V600 amplifications and BRAF splice site variants.99,118,126 Non-MAPK pathway alterations most frequently involve increased signaling through the PI3K pathway.99,101,127 Copy number variations in CDKN2A and CCND1 and inactivation of Rb have also been implicated in decreased response to BRAF/MEK inhibition although it is unclear whether this is due to more aggressive disease overall or a specific mechanistic link to resistance.128,129

Overcoming BRAF/MEK Inhibitor Resistance

Given the role of persistent MAPK signaling in resistance to BRAF/MEKi, inhibition farther downstream in the pathway was proposed as a potential mechanism to mitigate MAPK re-activation. Mitogen-activated extracellular-signal regulated kinase (ERK) is the final effector in the MAPK cascade and acts within the nucleus to promote proliferation, growth and survival. Pre-clinical studies have suggested that ERK inhibitors can independently induce regression in BRAF-mutant melanoma and may also reverse resistance to BRAF/MEKi.130 ERK inhibitors have produced responses in BRAF/MEKi resistant patients as well as treatment naïve BRAF and NRAS mutant melanomas in small early clinical trials.131,132

Re-introduction of BRAF/MEKi in patients who previously progressed on these agents has notably led to significant responses in some cases.133 Intermittent dosing of BRAFi has also induced more durable responses in mouse models compared with continuous dosing with regression of BRAF-amplified resistant tumors following BRAFi discontinuation.114,134 Evidence suggests that some BRAF/MEKi resistant melanomas may become “inhibitor addicted” and regress with short-term drug withdrawal.135 Intermittent dosing strategies may improve outcomes in these patients and are being evaluated in early phase trials (NCT02196181). However, one study recently presented negative results with a regimen of 5 weeks on and 3 weeks off dabrafenib and trametinib compared with continuous dosing.136

The AXL receptor is a target of interest in many malignancies, particularly in the setting of refractory or advanced disease.137 Several therapeutic agents are under investigation in early trials, including established multi-targeted kinase inhibitors shown to inhibit AXL and novel more specific small molecule inhibitors.138 AXL-directed antibody-drug-conjugates (ADC) have also been developed in an attempt to more specifically target AXL-expressing cell populations. When used in combination with BRAF/MEKi in patient-derived xenografts of melanoma with heterogeneous cell populations, an ADC containing the antimitotic agent monomethyl auristatin E (AXL-107-MMAE) eliminated tumor cells in the AXL-high population while BRAF/MEKi were effective in AXL-low cell lines. AXL-107-MMAE was also shown to potentiate the effect of BRAF/MEKi in AXL-low populations by exploiting BRAF/MEKi-induced transcriptional upregulation of AXL, suggesting a potential for benefit in BRAF/MEKi naïve patients. A phase I trial evaluating an AXL-specific ADC in advanced or relapsed/refractory solid tumors is ongoing (NCT02988817).

Inhibitors of heat shock protein-90 (HSP90), a chaperone that supports many RTKs and intracellular proteins involved in tumor growth and progression, have also been suggested as an adjunct therapy that may mitigate BRAF/MEKi resistance in melanoma. A phase trial I evaluating the HSP90 inhibitor XL888 and vemurafenib in BRAFi naïve patients with BRAF-mutant melanoma demonstrated a notable 75% response rate (15 of 20 evaluable patients) in addition to a tolerable toxicity profile.140 Multiple phase I trials are evaluating the use of these agents with dual BRAF/MEKi (NCT02721459, NCT02097225).

Key Points.

  1. Novel targeted therapies and immune checkpoint inhibitors have transformed the management of advanced melanoma, although resistance limits this benefit.

  2. Immune checkpoint inhibitors are often associated with intrinsic resistance and primary progressive disease, which may be due to lack of immune recognition, T cell exclusion, or alternative causes of T cell exhaustion.

  3. Targeted therapies are more often associated with acquired resistance, which may in part be due to reactivation of MAP kinase signaling and transcriptomic reprogramming.

Synopsis.

Novel therapeutic agents introduced over the last decade, including immune checkpoint inhibitors and targeted therapies, have revolutionized the management of metastatic melanoma and significantly improved patient outcomes. While robust and durable responses have been noted in some cases, treatment is often limited by innate or acquired resistance to these agents. This review provides an overview of known and suspected mechanisms involved with acquired resistance to BRAF/MEK inhibitors as well as developing insights into innate and acquired resistance to checkpoint inhibitors in patients with melanoma.

Disclosure Statement

  • Dr. April Salama receives research funding (paid to institution) from Bristol Myers Squibb, Immunocore, Merck and has served as a consultant/advisory board member for Array, Novartis, Iovance and Regeneron.

  • Dr. Douglas Johnson serves on advisory boards for Array Biopharma, BMS, Iovance, Jansen, Merck, and Novartis, and receives research funding from BMS and Incyte.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Contributor Information

Douglas B. Johnson, Department of Medicine, Vanderbilt University Medical Center, Vanderbilt Ingram Cancer Center, Nashville, TN, 777 PRB, 2220 Pierce Ave, Nashville, TN 37232.

April K.S. Salama, Department of Medicine, Duke University Medical Center.

Alexandra M. Haugh, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN.

References

  • 1.Yarchoan M, Hopkins A, Jaffee EM. Tumor Mutational Burden and Response Rate to PD-1 Inhibition. The New England journal of medicine. 2017;377(25):2500–2501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Samstein RM, Lee CH, Shoushtari AN, et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nature genetics. 2019;51(2):202–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Johnson DB, Frampton GM, Rioth MJ, et al. Targeted Next Generation Sequencing Identifies Markers of Response to PD-1 Blockade. Cancer Immunol Res. 2016;4(11):959–967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Van Allen EM, Miao D, Schilling B, et al. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science. 2015;350(6257):207–211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Rizvi NA, Hellmann MD, Snyder A, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348(6230):124–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Hellmann MD, Ciuleanu TE, Pluzanski A, et al. Nivolumab plus Ipilimumab in Lung Cancer with a High Tumor Mutational Burden. The New England journal of medicine. 2018;378(22):2093–2104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Snyder A, Makarov V, Merghoub T, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. The New England journal of medicine. 2014;371(23):2189–2199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.van Rooij N, van Buuren MM, Philips D, et al. Tumor exome analysis reveals neoantigen-specific T-cell reactivity in an ipilimumab-responsive melanoma. J Clin Oncol. 2013;31(32):e439–442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Le DT, Durham JN, Smith KN, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357(6349):409–413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Lipson EJ, Sharfman WH, Drake CG, et al. Durable cancer regression off-treatment and effective reinduction therapy with an anti-PD-1 antibody. Clin Cancer Res. 2013;19(2):462–468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Le DT, Uram JN, Wang H, et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. The New England journal of medicine. 2015;372(26):2509–2520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.You W, Shang B, Sun J, Liu X, Su L, Jiang S. Mechanistic insight of predictive biomarkers for antitumor PD‑1/PD‑L1 blockade: A paradigm shift towards immunome evaluation (Review). Oncol Rep. 2020;44(2):424–437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Miao D, Margolis CA, Vokes NI, et al. Genomic correlates of response to immune checkpoint blockade in microsatellite-stable solid tumors. Nature genetics. 2018;50(9):1271–1281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.McGranahan N, Furness AJ, Rosenthal R, et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science. 2016;351(6280):1463–1469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Hayward NK, Wilmott JS, Waddell N, et al. Whole-genome landscapes of major melanoma subtypes. Nature. 2017;545(7653):175–180. [DOI] [PubMed] [Google Scholar]
  • 16.Shoushtari AN, Munhoz RR, Kuk D, et al. The efficacy of anti-PD-1 agents in acral and mucosal melanoma. Cancer. 2016;122(21):3354–3362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Johnson DB, Peng C, Abramson RG, et al. Clinical Activity of Ipilimumab in Acral Melanoma: A Retrospective Review. Oncologist. 2015;20(6):648–652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Nakamura Y, Namikawa K, Yoshino K, et al. Anti-PD1 checkpoint inhibitor therapy in acral melanoma: a multicenter study of 193 Japanese patients. Ann Oncol. 2020. [DOI] [PubMed] [Google Scholar]
  • 19.Eroglu Z, Zaretsky JM, Hu-Lieskovan S, et al. High response rate to PD-1 blockade in desmoplastic melanomas. Nature. 2018;553(7688):347–350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Verdegaal EM, de Miranda NF, Visser M, et al. Neoantigen landscape dynamics during human melanoma-T cell interactions. Nature. 2016;536(7614):91–95. [DOI] [PubMed] [Google Scholar]
  • 21.Matsushita H, Vesely MD, Koboldt DC, et al. Cancer exome analysis reveals a T-cell-dependent mechanism of cancer immunoediting. Nature. 2012;482(7385):400–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Anagnostou V, Smith KN, Forde PM, et al. Evolution of Neoantigen Landscape during Immune Checkpoint Blockade in Non-Small Cell Lung Cancer. Cancer Discov. 2017;7(3):264–276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. The New England journal of medicine. 2012;366(26):2443–2454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Weber JS, Kudchadkar RR, Yu B, et al. Safety, efficacy, and biomarkers of nivolumab with vaccine in ipilimumab-refractory or -naive melanoma. J Clin Oncol. 2013;31(34):4311–4318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Larkin J, Chiarion-Sileni V, Gonzalez R, et al. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. The New England journal of medicine. 2015;373(1):23–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Robert C, Long GV, Brady B, et al. Nivolumab in previously untreated melanoma without BRAF mutation. The New England journal of medicine. 2015;372(4):320–330. [DOI] [PubMed] [Google Scholar]
  • 27.Daud AI, Loo K, Pauli ML, et al. Tumor immune profiling predicts response to anti-PD-1 therapy in human melanoma. The Journal of clinical investigation. 2016;126(9):3447–3452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Taube JM, Klein A, Brahmer JR, et al. Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin Cancer Res. 2014;20(19):5064–5074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Bald T, Landsberg J, Lopez-Ramos D, et al. Immune cell-poor melanomas benefit from PD-1 blockade after targeted type I IFN activation. Cancer Discov. 2014;4(6):674–687. [DOI] [PubMed] [Google Scholar]
  • 30.Rooney MS, Shukla SA, Wu CJ, Getz G, Hacohen N. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell. 2015;160(1–2):48–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Zhao F, Sucker A, Horn S, et al. Melanoma Lesions Independently Acquire T-cell Resistance during Metastatic Latency. Cancer research. 2016;76(15):4347–4358. [DOI] [PubMed] [Google Scholar]
  • 32.Zaretsky JM, Garcia-Diaz A, Shin DS, et al. Mutations Associated with Acquired Resistance to PD-1 Blockade in Melanoma. The New England journal of medicine. 2016;375(9):819–829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Rodig SJ, Gusenleitner D, Jackson DG, et al. MHC proteins confer differential sensitivity to CTLA-4 and PD-1 blockade in untreated metastatic melanoma. Sci Transl Med. 2018;10(450). [DOI] [PubMed] [Google Scholar]
  • 34.Chowell D, Morris LGT, Grigg CM, et al. Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science. 2018;359(6375):582–587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Shin DS, Zaretsky JM, Escuin-Ordinas H, et al. Primary Resistance to PD-1 Blockade Mediated by JAK1/2 Mutations. Cancer Discov. 2017;7(2):188–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Gao J, Shi LZ, Zhao H, et al. Loss of IFN-γ Pathway Genes in Tumor Cells as a Mechanism of Resistance to Anti-CTLA-4 Therapy. Cell. 2016;167(2):397–404. e399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature. 2015;523(7559):231–235. [DOI] [PubMed] [Google Scholar]
  • 38.Spranger S, Luke JJ, Bao R, et al. Density of immunogenic antigens does not explain the presence or absence of the T-cell-inflamed tumor microenvironment in melanoma. Proceedings of the National Academy of Sciences of the United States of America. 2016;113(48):E7759–e7768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Sade-Feldman M, Yizhak K, Bjorgaard SL, et al. Defining T Cell States Associated with Response to Checkpoint Immunotherapy in Melanoma. Cell. 2018;175(4):998–1013. e1020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Peng W, Chen JQ, Liu C, et al. Loss of PTEN Promotes Resistance to T Cell-Mediated Immunotherapy. Cancer Discov. 2016;6(2):202–216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.George S, Miao D, Demetri GD, et al. Loss of PTEN Is Associated with Resistance to Anti-PD-1 Checkpoint Blockade Therapy in Metastatic Uterine Leiomyosarcoma. Immunity. 2017;46(2):197–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Boni A, Cogdill AP, Dang P, et al. Selective BRAFV600E inhibition enhances T-cell recognition of melanoma without affecting lymphocyte function. Cancer research. 2010;70(13):5213–5219. [DOI] [PubMed] [Google Scholar]
  • 43.Liu C, Peng W, Xu C, et al. BRAF inhibition increases tumor infiltration by T cells and enhances the antitumor activity of adoptive immunotherapy in mice. Clin Cancer Res. 2013;19(2):393–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Frederick DT, Piris A, Cogdill AP, et al. BRAF inhibition is associated with enhanced melanoma antigen expression and a more favorable tumor microenvironment in patients with metastatic melanoma. Clin Cancer Res. 2013;19(5):1225–1231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Schilling B, Paschen A. Immunological consequences of selective BRAF inhibitors in malignant melanoma: Neutralization of myeloid-derived suppressor cells. Oncoimmunology. 2013;2(8):e25218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Wilmott JS, Long GV, Howle JR, et al. Selective BRAF inhibitors induce marked T-cell infiltration into human metastatic melanoma. Clin Cancer Res. 2012;18(5):1386–1394. [DOI] [PubMed] [Google Scholar]
  • 47.Liu L, Mayes PA, Eastman S, 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):1639–1651. [DOI] [PubMed] [Google Scholar]
  • 48.Cooper ZA, Reuben A, Spencer CN, et al. Distinct clinical patterns and immune infiltrates are observed at time of progression on targeted therapy versus immune checkpoint blockade for melanoma. Oncoimmunology. 2016;5(3):e1136044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Hugo W, Zaretsky JM, Sun L, et al. Genomic and Transcriptomic Features of Response to Anti-PD-1 Therapy in Metastatic Melanoma. Cell. 2017;168(3):542. [DOI] [PubMed] [Google Scholar]
  • 50.Yan Y, Wongchenko MJ, Robert C, et al. Genomic Features of Exceptional Response in Vemurafenib +/− Cobimetinib-treated Patients with BRAF (V600)-mutated Metastatic Melanoma. Clin Cancer Res. 2019;25(11):3239–3246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Johnson DB, Pectasides E, Feld E, et al. Sequencing Treatment in BRAFV600 Mutant Melanoma: Anti-PD-1 Before and After BRAF Inhibition. J Immunother. 2017;40(1):31–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Johnson DB, Lovly CM, Flavin M, et al. Impact of NRAS mutations for patients with advanced melanoma treated with immune therapies. Cancer Immunol Res. 2015;3(3):288–295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Fridman WH, Pagès F, Sautès-Fridman C, Galon J. The immune contexture in human tumours: impact on clinical outcome. Nature reviews Cancer. 2012;12(4):298–306. [DOI] [PubMed] [Google Scholar]
  • 54.Tumeh PC, Harview CL, Yearley JH, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515(7528):568–571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Wherry EJ, Kurachi M. Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol. 2015;15(8):486–499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Pauken KE, Sammons MA, Odorizzi PM, et al. Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade. Science. 2016;354(6316):1160–1165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Sen DR, Kaminski J, Barnitz RA, et al. The epigenetic landscape of T cell exhaustion. Science. 2016;354(6316):1165–1169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Mognol GP, Spreafico R, Wong V, et al. Exhaustion-associated regulatory regions in CD8(+) tumor-infiltrating T cells. Proceedings of the National Academy of Sciences of the United States of America. 2017;114(13):E2776–e2785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Francisco LM, Salinas VH, Brown KE, et al. PD-L1 regulates the development, maintenance, and function of induced regulatory T cells. J Exp Med. 2009;206(13):3015–3029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Ding ZC, Lu X, Yu M, et al. Immunosuppressive myeloid cells induced by chemotherapy attenuate antitumor CD4+ T-cell responses through the PD-1-PD-L1 axis. Cancer research. 2014;74(13):3441–3453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Avogadri F, Zappasodi R, Yang A, et al. Combination of alphavirus replicon particle-based vaccination with immunomodulatory antibodies: therapeutic activity in the B16 melanoma mouse model and immune correlates. Cancer Immunol Res. 2014;2(5):448–458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Zappasodi R, Budhu S, Hellmann MD, et al. Non-conventional Inhibitory CD4(+)Foxp3(−)PD-1(hi) T Cells as a Biomarker of Immune Checkpoint Blockade Activity. Cancer cell. 2018;33(6):1017–1032. e1017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Hugo W, Zaretsky JM, Sun L, et al. Genomic and Transcriptomic Features of Response to Anti-PD-1 Therapy in Metastatic Melanoma. Cell. 2016;165(1):35–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Chen L, Gibbons DL, Goswami S, et al. Metastasis is regulated via microRNA-200/ZEB1 axis control of tumour cell PD-L1 expression and intratumoral immunosuppression. Nat Commun. 2014;5:5241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Chen L, Heymach JV, Qin FX, Gibbons DL. The mutually regulatory loop of epithelial-mesenchymal transition and immunosuppression in cancer progression. Oncoimmunology. 2015;4(5):e1002731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Mak MP, Tong P, Diao L, et al. A Patient-Derived, Pan-Cancer EMT Signature Identifies Global Molecular Alterations and Immune Target Enrichment Following Epithelial-to-Mesenchymal Transition. Clin Cancer Res. 2016;22(3):609–620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Falletta P, Sanchez-Del-Campo L, Chauhan J, et al. Translation reprogramming is an evolutionarily conserved driver of phenotypic plasticity and therapeutic resistance in melanoma. Genes Dev. 2017;31(1):18–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Minn AJ, Wherry EJ. Combination Cancer Therapies with Immune Checkpoint Blockade: Convergence on Interferon Signaling. Cell. 2016;165(2):272–275. [DOI] [PubMed] [Google Scholar]
  • 69.Herbst RS, Soria JC, Kowanetz M, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515(7528):563–567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Ayers M, Lunceford J, Nebozhyn M, et al. IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. The Journal of clinical investigation. 2017;127(8):2930–2940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Benci JL, Xu B, Qiu Y, et al. Tumor Interferon Signaling Regulates a Multigenic Resistance Program to Immune Checkpoint Blockade. Cell. 2016;167(6):1540–1554. e1512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Takeda K, Nakayama M, Hayakawa Y, et al. IFN-γ is required for cytotoxic T cell-dependent cancer genome immunoediting. Nat Commun. 2017;8:14607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Harel M, Ortenberg R, Varanasi SK, et al. Proteomics of Melanoma Response to Immunotherapy Reveals Mitochondrial Dependence. Cell. 2019;179(1):236–250. e218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Gopalakrishnan V, Spencer CN, Nezi L, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science. 2018;359(6371):97–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Chaput N, Lepage P, Coutzac C, et al. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann Oncol. 2017;28(6):1368–1379. [DOI] [PubMed] [Google Scholar]
  • 76.Matson V, Fessler J, Bao R, et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science. 2018;359(6371):104–108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Pinato DJ, Howlett S, Ottaviani D, et al. Association of Prior Antibiotic Treatment With Survival and Response to Immune Checkpoint Inhibitor Therapy in Patients With Cancer. JAMA Oncol. 2019;5(12):1774–1778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Routy B, Le Chatelier E, Derosa L, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;359(6371):91–97. [DOI] [PubMed] [Google Scholar]
  • 79.Benus RF, van der Werf TS, Welling GW, et al. Association between Faecalibacterium prausnitzii and dietary fibre in colonic fermentation in healthy human subjects. Br J Nutr. 2010;104(5):693–700. [DOI] [PubMed] [Google Scholar]
  • 80.Spencer CN, Gopalakrishnan V, McQuade JL. The gut microbiome (GM) and immunotherapy response are influenced by host lifestyle factors. AACR Annual Meeting. April 2019. [Google Scholar]
  • 81.Twyman-Saint Victor C, Rech AJ, Maity A, et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature. 2015;520(7547):373–377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Maurer DM, Butterfield LH, Vujanovic L. Melanoma vaccines: clinical status and immune endpoints. Melanoma research. 2019;29(2):109–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Zamarin D, Holmgaard RB, Subudhi SK, et al. Localized oncolytic virotherapy overcomes systemic tumor resistance to immune checkpoint blockade immunotherapy. Sci Transl Med. 2014;6(226):226ra232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Fu J, Kanne DB, Leong M, et al. STING agonist formulated cancer vaccines can cure established tumors resistant to PD-1 blockade. Sci Transl Med. 2015;7(283):283ra252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Fares CM, Van Allen EM, Drake CG, Allison JP, Hu-Lieskovan S. Mechanisms of Resistance to Immune Checkpoint Blockade: Why Does Checkpoint Inhibitor Immunotherapy Not Work for All Patients? Am Soc Clin Oncol Educ Book 2019;39:147–164. [DOI] [PubMed] [Google Scholar]
  • 86.Jerby-Arnon L, Shah P, Cuoco MS, et al. A Cancer Cell Program Promotes T Cell Exclusion and Resistance to Checkpoint Blockade. Cell. 2018;175(4):984–997. e924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Ascierto P, Melero I, Bhatia S et al. Initial efficacy of anti-lymphocyte activation gene-3 (anti–LAG-3; BMS-986016) in combination with nivolumab (nivo) in pts with melanoma (MEL) previously treated with anti–PD-1/PD-L1 therapy. Journal of Clinical Oncology.35:9520–9520. [Google Scholar]
  • 88.Reilley MJ, Morrow B, Ager CR, Liu A, Hong DS, Curran MA. TLR9 activation cooperates with T cell checkpoint blockade to regress poorly immunogenic melanoma. J Immunother Cancer. 2019;7(1):323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Diab ARS, Haymaker CL. A phase 2 study to evaluate the safety and efficacy of Intratumoral (IT) injection of the TLR9 agonist IMO-2125 (IMO) in combination with ipilimumab (ipi) in PD-1 inhibitor refractory melanoma. Paper presented at: 2018 Annual ASCO Meeting; 2018; Chicago, IL [Google Scholar]
  • 90.Sarnaik AKN, Chesney JA. Long-term follow up of lifileucel (LN-144) cryopreserved autologous tumor infiltrating lymphocyte therapy in patients with advanced melanoma progressed on multiple prior therapies.;38. [Google Scholar]
  • 91.Chapman PB, Robert C, Larkin J, et al. Vemurafenib in patients with BRAFV600 mutation-positive metastatic melanoma: final overall survival results of the randomized BRIM-3 study. Ann Oncol. 2017;28(10):2581–2587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Hauschild A, Grob JJ, Demidov LV, et al. An update on BREAK-3, a phase III, randomized trial: Dabrafenib (DAB) versus dacarbazine (DTIC) in patients with BRAF V600E-positive mutation metastatic melanoma (MM). Journal of Clinical Oncology. 2013;31(15_suppl):9013–9013. [Google Scholar]
  • 93.Flaherty KT, Robert C, Hersey P, et al. Improved survival with MEK inhibition in BRAF-mutated melanoma. The New England journal of medicine. 2012;367(2):107–114. [DOI] [PubMed] [Google Scholar]
  • 94.Dummer R, Ascierto PA, Gogas HJ, et al. Overall survival in patients with BRAF-mutant melanoma receiving encorafenib plus binimetinib versus vemurafenib or encorafenib (COLUMBUS): a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2018;19(10):1315–1327. [DOI] [PubMed] [Google Scholar]
  • 95.Ascierto PA, McArthur GA, Dreno B, et al. Cobimetinib combined with vemurafenib in advanced BRAF(V600)-mutant melanoma (coBRIM): updated efficacy results from a randomised, double-blind, phase 3 trial. Lancet Oncol. 2016;17(9):1248–1260. [DOI] [PubMed] [Google Scholar]
  • 96.Larkin J, Ascierto PA, Dreno B, et al. Combined vemurafenib and cobimetinib in BRAF-mutated melanoma. The New England journal of medicine. 2014;371(20):1867–1876. [DOI] [PubMed] [Google Scholar]
  • 97.Wagle N, Van Allen EM, Treacy DJ, et al. MAP kinase pathway alterations in BRAF-mutant melanoma patients with acquired resistance to combined RAF/MEK inhibition. Cancer Discov. 2014;4(1):61–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Long GV, Fung C, Menzies AM, et al. Increased MAPK reactivation in early resistance to dabrafenib/trametinib combination therapy of BRAF-mutant metastatic melanoma. Nat Commun. 2014;5:5694. [DOI] [PubMed] [Google Scholar]
  • 99.Johnson DB, Menzies AM, Zimmer L, et al. Acquired BRAF inhibitor resistance: A multicenter meta-analysis of the spectrum and frequencies, clinical behaviour, and phenotypic associations of resistance mechanisms. Eur J Cancer. 2015;51(18):2792–2799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Robert C, Grob JJ, Stroyakovskiy D, et al. Five-Year Outcomes with Dabrafenib plus Trametinib in Metastatic Melanoma. The New England journal of medicine. 2019. [DOI] [PubMed] [Google Scholar]
  • 101.Shi H, Hugo W, Kong X, et al. Acquired resistance and clonal evolution in melanoma during BRAF inhibitor therapy. Cancer Discov. 2014;4(1):80–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Ravindran Menon D, Das S, Krepler C, et al. A stress-induced early innate response causes multidrug tolerance in melanoma. Oncogene. 2015;34(34):4448–4459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Webster MR, Xu M, Kinzler KA, et al. Wnt5A promotes an adaptive, senescent-like stress response, while continuing to drive invasion in melanoma cells. Pigment Cell Melanoma Res. 2015;28(2):184–195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Sun C, Wang L, Huang S, et al. Reversible and adaptive resistance to BRAF(V600E) inhibition in melanoma. Nature. 2014;508(7494):118–122. [DOI] [PubMed] [Google Scholar]
  • 105.Hugo W, Shi H, Sun L, et al. Non-genomic and Immune Evolution of Melanoma Acquiring MAPKi Resistance. Cell. 2015;162(6):1271–1285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Titz B, Lomova A, Le A, et al. JUN dependency in distinct early and late BRAF inhibition adaptation states of melanoma. Cell Discov. 2016;2:16028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Fedorenko IV, Abel EV, Koomen JM, et al. Fibronectin induction abrogates the BRAF inhibitor response of BRAF V600E/PTEN-null melanoma cells. Oncogene. 2016;35(10):1225–1235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Ramsdale R, Jorissen RN, Li FZ, et al. The transcription cofactor c-JUN mediates phenotype switching and BRAF inhibitor resistance in melanoma. Sci Signal. 2015;8(390):ra82. [DOI] [PubMed] [Google Scholar]
  • 109.Fallahi-Sichani M, Moerke NJ, Niepel M, Zhang T, Gray NS, Sorger PK. Systematic analysis of BRAF(V600E) melanomas reveals a role for JNK/c-Jun pathway in adaptive resistance to drug-induced apoptosis. Mol Syst Biol. 2015;11(3):797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Konieczkowski DJ, Johannessen CM, Abudayyeh O, et al. A melanoma cell state distinction influences sensitivity to MAPK pathway inhibitors. Cancer Discov. 2014;4(7):816–827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Müller J, Krijgsman O, Tsoi J, et al. Low MITF/AXL ratio predicts early resistance to multiple targeted drugs in melanoma. Nat Commun. 2014;5:5712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Johannessen CM, Johnson LA, Piccioni F, et al. A melanocyte lineage program confers resistance to MAP kinase pathway inhibition. Nature. 2013;504(7478):138–142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Wellbrock C, Arozarena I. Microphthalmia-associated transcription factor in melanoma development and MAP-kinase pathway targeted therapy. Pigment Cell Melanoma Res. 2015;28(4):390–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Lim SY, Menzies AM, Rizos H. Mechanisms and strategies to overcome resistance to molecularly targeted therapy for melanoma. Cancer. 2017;123(S11):2118–2129. [DOI] [PubMed] [Google Scholar]
  • 115.Catalanotti F, Solit DB, Pulitzer MP, et al. Phase II trial of MEK inhibitor selumetinib (AZD6244, ARRY-142886) in patients with BRAFV600E/K-mutated melanoma. Clin Cancer Res. 2013;19(8):2257–2264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Gopal YN, Deng W, Woodman SE, et al. Basal and treatment-induced activation of AKT mediates resistance to cell death by AZD6244 (ARRY-142886) in Braf-mutant human cutaneous melanoma cells. Cancer research. 2010;70(21):8736–8747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Ascierto PA, Flaherty K, Goff S. Emerging Strategies in Systemic Therapy for the Treatment of Melanoma. Am Soc Clin Oncol Educ Book. 2018;38:751–758. [DOI] [PubMed] [Google Scholar]
  • 118.Villanueva J, Vultur A, Lee JT, et al. Acquired resistance to BRAF inhibitors mediated by a RAF kinase switch in melanoma can be overcome by cotargeting MEK and IGF-1R/PI3K. Cancer cell. 2010;18(6):683–695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Abel EV, Basile KJ, Kugel CH 3rd, et al. Melanoma adapts to RAF/MEK inhibitors through FOXD3-mediated upregulation of ERBB3. The Journal of clinical investigation. 2013;123(5):2155–2168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Nazarian R, Shi H, Wang Q, et al. Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation. Nature. 2010;468(7326):973–977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Girotti MR, Pedersen M, Sanchez-Laorden B, et al. Inhibiting EGF receptor or SRC family kinase signaling overcomes BRAF inhibitor resistance in melanoma. Cancer Discov. 2013;3(2):158–167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Paraiso KH, Fedorenko IV, Cantini LP, et al. Recovery of phospho-ERK activity allows melanoma cells to escape from BRAF inhibitor therapy. British journal of cancer. 2010;102(12):1724–1730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Straussman R, Morikawa T, Shee K, et al. Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature. 2012;487(7408):500–504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Sensi M, Catani M, Castellano G, et al. Human cutaneous melanomas lacking MITF and melanocyte differentiation antigens express a functional Axl receptor kinase. The Journal of investigative dermatology. 2011;131(12):2448–2457. [DOI] [PubMed] [Google Scholar]
  • 125.Rizos H, Menzies AM, Pupo GM, et al. BRAF inhibitor resistance mechanisms in metastatic melanoma: spectrum and clinical impact. Clin Cancer Res. 2014;20(7):1965–1977. [DOI] [PubMed] [Google Scholar]
  • 126.Wagle N, Emery C, Berger MF, et al. Dissecting therapeutic resistance to RAF inhibition in melanoma by tumor genomic profiling. J Clin Oncol. 2011;29(22):3085–3096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Shi H, Hong A, Kong X, et al. A novel AKT1 mutant amplifies an adaptive melanoma response to BRAF inhibition. Cancer Discov. 2014;4(1):69–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Xing F, Persaud Y, Pratilas CA, et al. Concurrent loss of the PTEN and RB1 tumor suppressors attenuates RAF dependence in melanomas harboring (V600E)BRAF. Oncogene. 2012;31(4):446–457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Smalley KS, Lioni M, Dalla Palma M, et al. Increased cyclin D1 expression can mediate BRAF inhibitor resistance in BRAF V600E-mutated melanomas. Mol Cancer Ther. 2008;7(9):2876–2883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Morris EJ, Jha S, Restaino CR, et al. Discovery of a novel ERK inhibitor with activity in models of acquired resistance to BRAF and MEK inhibitors. Cancer Discov. 2013;3(7):742–750. [DOI] [PubMed] [Google Scholar]
  • 131.Sullivan RJ, Infante JR, Janku F, et al. First-in-Class ERK1/2 Inhibitor Ulixertinib (BVD-523) in Patients with MAPK Mutant Advanced Solid Tumors: Results of a Phase I Dose-Escalation and Expansion Study. Cancer Discov. 2018;8(2):184–195. [DOI] [PubMed] [Google Scholar]
  • 132.Moschos SJ, Sullivan RJ, Hwu WJ, et al. Development of MK-8353, an orally administered ERK1/2 inhibitor, in patients with advanced solid tumors. JCI Insight. 2018;3(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Schreuer M, Jansen Y, Planken S, et al. Combination of dabrafenib plus trametinib for BRAF and MEK inhibitor pretreated patients with advanced BRAF(V600)-mutant melanoma: an open-label, single arm, dual-centre, phase 2 clinical trial. Lancet Oncol. 2017;18(4):464–472. [DOI] [PubMed] [Google Scholar]
  • 134.Das Thakur M, Salangsang F, Landman AS, et al. Modelling vemurafenib resistance in melanoma reveals a strategy to forestall drug resistance. Nature. 2013;494(7436):251–255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Moriceau G, Hugo W, Hong A, et al. Tunable-combinatorial mechanisms of acquired resistance limit the efficacy of BRAF/MEK cotargeting but result in melanoma drug addiction. Cancer cell. 2015;27(2):240–256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Algazi A O M, Daud A et al. CT013 - SWOG S1320: Improved progression-free survival with continuous compared to intermittent dosing with dabrafenib and trametinib in patients with BRAF mutated melanoma. Paper presented at: American Association for Cancer Research (AACR) Virtual Annual Meeting; April 27, 2020. [Google Scholar]
  • 137.Gay CM, Balaji K, Byers LA. Giving AXL the axe: targeting AXL in human malignancy. British journal of cancer. 2017;116(4):415–423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Zhu C, Wei Y, Wei X. AXL receptor tyrosine kinase as a promising anti-cancer approach: functions, molecular mechanisms and clinical applications. Mol Cancer. 2019;18(1):153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Boshuizen J, Koopman LA, Krijgsman O, et al. Cooperative targeting of melanoma heterogeneity with an AXL antibody-drug conjugate and BRAF/MEK inhibitors. Nat Med. 2018;24(2):203–212. [DOI] [PubMed] [Google Scholar]
  • 140.Eroglu Z, Chen YA, Gibney GT, et al. Combined BRAF and HSP90 Inhibition in Patients with Unresectable BRAF (V600E)-Mutant Melanoma. Clin Cancer Res. 2018;24(22):5516–5524. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES