Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Nat Commun ; 11(1): 2345, 2020 05 11.
Article in English | MEDLINE | ID: mdl-32393797

ABSTRACT

The determination of individual cell trajectories through a high-dimensional cell-state space is an outstanding challenge for understanding biological changes ranging from cellular differentiation to epigenetic responses of diseased cells upon drugging. We integrate experiments and theory to determine the trajectories that single BRAFV600E mutant melanoma cancer cells take between drug-naive and drug-tolerant states. Although single-cell omics tools can yield snapshots of the cell-state landscape, the determination of individual cell trajectories through that space can be confounded by stochastic cell-state switching. We assayed for a panel of signaling, phenotypic, and metabolic regulators at points across 5 days of drug treatment to uncover a cell-state landscape with two paths connecting drug-naive and drug-tolerant states. The trajectory a given cell takes depends upon the drug-naive level of a lineage-restricted transcription factor. Each trajectory exhibits unique druggable susceptibilities, thus updating the paradigm of adaptive resistance development in an isogenic cell population.


Subject(s)
Drug Tolerance , Genomics , Melanoma/drug therapy , Single-Cell Analysis , Cell Line, Tumor , Genes, Reporter , Green Fluorescent Proteins/metabolism , Humans , Metabolomics , Microphthalmia-Associated Transcription Factor , Models, Molecular , Proteomics , Proto-Oncogene Proteins B-raf/genetics , Reproducibility of Results
2.
PLoS Comput Biol ; 15(6): e1007034, 2019 06.
Article in English | MEDLINE | ID: mdl-31166947

ABSTRACT

Phenotypic plasticity is associated with non-genetic drug tolerance in several cancers. Such plasticity can arise from chromatin remodeling, transcriptomic reprogramming, and/or protein signaling rewiring, and is characterized as a cell state transition in response to molecular or physical perturbations. This, in turn, can confound interpretations of drug responses and resistance development. Using BRAF-mutant melanoma cell lines as the prototype, we report on a joint theoretical and experimental investigation of the cell-state transition dynamics associated with BRAF inhibitor drug tolerance. Thermodynamically motivated surprisal analysis of transcriptome data was used to treat the cell population as an entropy maximizing system under the influence of time-dependent constraints. This permits the extraction of an epigenetic potential landscape for drug-induced phenotypic evolution. Single-cell flow cytometry data of the same system were modeled with a modified Fokker-Planck-type kinetic model. The two approaches yield a consistent picture that accounts for the phenotypic heterogeneity observed over the course of drug tolerance development. The results reveal that, in certain plastic cancers, the population heterogeneity and evolution of cell phenotypes may be understood by accounting for the competing interactions of the epigenetic potential landscape and state-dependent cell proliferation. Accounting for such competition permits accurate, experimentally verifiable predictions that can potentially guide the design of effective treatment strategies.


Subject(s)
Drug Resistance, Neoplasm , Evolution, Molecular , Melanoma , Phenotype , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Humans , Melanoma/genetics , Melanoma/physiopathology , Models, Biological , Transcriptome/drug effects , Transcriptome/genetics
3.
Cancer Discov ; 8(8): 935-943, 2018 08.
Article in English | MEDLINE | ID: mdl-29899062

ABSTRACT

A promising arsenal of targeted and immunotherapy treatments for metastatic melanoma has emerged over the last decade. With these therapies, we now face new mechanisms of tumor-acquired resistance. We report here a patient whose metastatic melanoma underwent dedifferentiation as a resistance mechanism to adoptive T-cell transfer therapy (ACT) to the MART1 antigen, a phenomenon that had been observed only in mouse studies to date. After an initial period of tumor regression, the patient presented in relapse with tumors lacking melanocytic antigens (MART1, gp100) and expressing an inflammation-induced neural crest marker (NGFR). We demonstrate using human melanoma cell lines that this resistance phenotype can be induced in vitro by treatment with MART1 T cell receptor-expressing T cells or with TNFα, and that the phenotype is reversible with withdrawal of inflammatory stimuli. This supports the hypothesis that acquired resistance to cancer immunotherapy can be mediated by inflammation-induced cancer dedifferentiation.Significance: We report a patient whose metastatic melanoma underwent inflammation-induced dedifferentiation as a resistance mechanism to ACT to the MART1 antigen. Our results suggest that future melanoma ACT protocols may benefit from the simultaneous targeting of multiple tumor antigens, modulating the inflammatory response, and inhibition of inflammatory dedifferentiation-inducing signals. Cancer Discov; 8(8); 935-43. ©2018 AACR.This article is highlighted in the In This Issue feature, p. 899.


Subject(s)
Drug Resistance, Neoplasm , MART-1 Antigen/immunology , Melanoma/therapy , Nerve Tissue Proteins/metabolism , Nevus, Pigmented/therapy , Receptors, Nerve Growth Factor/metabolism , Cell Dedifferentiation , Cell Line, Tumor , Coculture Techniques , Humans , Immunotherapy, Adoptive , Male , Melanoma/immunology , Middle Aged , Neoplasm Metastasis , Nevus, Pigmented/immunology , Receptors, Chimeric Antigen/metabolism , Recurrence
4.
Cancer Cell ; 33(5): 890-904.e5, 2018 05 14.
Article in English | MEDLINE | ID: mdl-29657129

ABSTRACT

Malignant transformation can result in melanoma cells that resemble different stages of their embryonic development. Our gene expression analysis of human melanoma cell lines and patient tumors revealed that melanoma follows a two-dimensional differentiation trajectory that can be subclassified into four progressive subtypes. This differentiation model is associated with subtype-specific sensitivity to iron-dependent oxidative stress and cell death known as ferroptosis. Receptor tyrosine kinase-mediated resistance to mitogen-activated protein kinase targeted therapies and activation of the inflammatory signaling associated with immune therapy involves transitions along this differentiation trajectory, which lead to increased sensitivity to ferroptosis. Therefore, ferroptosis-inducing drugs present an orthogonal therapeutic approach to target the differentiation plasticity of melanoma cells to increase the efficacy of targeted and immune therapies.


Subject(s)
Gene Expression Profiling/methods , Iron/metabolism , Melanoma/classification , Melanoma/genetics , Vemurafenib/pharmacology , Cell Dedifferentiation , Cell Line, Tumor , Cell Survival/drug effects , DNA Methylation , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Regulatory Networks , Humans , Iron/toxicity , Melanoma/drug therapy , Melanoma/metabolism , Oxidative Stress/drug effects , Piperazines , Signal Transduction
5.
JCI Insight ; 3(4)2018 02 22.
Article in English | MEDLINE | ID: mdl-29467321

ABSTRACT

BACKGROUND: Constitutive activation of ERK1/2 occurs in various cancers, and its reactivation is a well-described resistance mechanism to MAPK inhibitors. ERK inhibitors may overcome the limitations of MAPK inhibitor blockade. The dual mechanism inhibitor SCH772984 has shown promising preclinical activity across various BRAFV600/RAS-mutant cancer cell lines and human cancer xenografts. METHODS: We have developed an orally bioavailable ERK inhibitor, MK-8353; conducted preclinical studies to demonstrate activity, pharmacodynamic endpoints, dosing, and schedule; completed a study in healthy volunteers (P07652); and subsequently performed a phase I clinical trial in patients with advanced solid tumors (MK-8353-001). In the P07652 study, MK-8353 was administered as a single dose in 10- to 400-mg dose cohorts, whereas in the MK-8353-001 study, MK-8353 was administered in 100- to 800-mg dose cohorts orally twice daily. Safety, tolerability, pharmacokinetics, pharmacodynamics, and antitumor activity were analyzed. RESULTS: MK-8353 exhibited comparable potency with SCH772984 across various preclinical cancer models. Forty-eight patients were enrolled in the P07652 study, and twenty-six patients were enrolled in the MK-8353-001 study. Adverse events included diarrhea (44%), fatigue (40%), nausea (32%), and rash (28%). Dose-limiting toxicity was observed in the 400-mg and 800-mg dose cohorts. Sufficient exposure to MK-8353 was noted that correlated with biological activity in preclinical data. Three of fifteen patients evaluable for treatment response in the MK-8353-001 study had partial response, all with BRAFV600-mutant melanomas. CONCLUSION: MK-8353 was well tolerated up to 400 mg twice daily and exhibited antitumor activity in patients with BRAFV600-mutant melanoma. However, antitumor activity was not particularly correlated with pharmacodynamic parameters. TRIAL REGISTRATION: ClinicalTrials.gov NCT01358331. FUNDING: Merck Sharp & Dohme Corp., a subsidiary of Merck & Co. Inc., and NIH (P01 CA168585 and R35 CA197633).


Subject(s)
Indazoles/pharmacology , MAP Kinase Signaling System/drug effects , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Pyrrolidines/pharmacology , Triazoles/pharmacology , Administration, Oral , Adult , Animals , Biological Availability , Cell Line, Tumor , Diarrhea/chemically induced , Diarrhea/epidemiology , Dogs , Dose-Response Relationship, Drug , Drug Eruptions/epidemiology , Drug Eruptions/etiology , Drug Evaluation, Preclinical , Fatigue/chemically induced , Fatigue/epidemiology , Female , Humans , Indazoles/therapeutic use , Male , Maximum Tolerated Dose , Mice , Middle Aged , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Nausea/chemically induced , Nausea/epidemiology , Neoplasm Staging , Neoplasms/genetics , Neoplasms/pathology , Protein Kinase Inhibitors/therapeutic use , Pyridines/therapeutic use , Pyrrolidines/therapeutic use , Rats , Triazoles/therapeutic use , Xenograft Model Antitumor Assays , Young Adult
6.
Proc Natl Acad Sci U S A ; 114(52): 13679-13684, 2017 12 26.
Article in English | MEDLINE | ID: mdl-29229836

ABSTRACT

Continuous BRAF inhibition of BRAF mutant melanomas triggers a series of cell state changes that lead to therapy resistance and escape from immune control before establishing acquired resistance genetically. We used genome-wide transcriptomics and single-cell phenotyping to explore the response kinetics to BRAF inhibition for a panel of patient-derived BRAFV600 -mutant melanoma cell lines. A subset of plastic cell lines, which followed a trajectory covering multiple known cell state transitions, provided models for more detailed biophysical investigations. Markov modeling revealed that the cell state transitions were reversible and mediated by both Lamarckian induction and nongenetic Darwinian selection of drug-tolerant states. Single-cell functional proteomics revealed activation of certain signaling networks shortly after BRAF inhibition, and before the appearance of drug-resistant phenotypes. Drug targeting those networks, in combination with BRAF inhibition, halted the adaptive transition and led to prolonged growth inhibition in multiple patient-derived cell lines.


Subject(s)
Drug Resistance, Neoplasm , Melanoma/genetics , Melanoma/metabolism , Signal Transduction , Single-Cell Analysis , Adaptation, Physiological , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Gene Expression Profiling , Humans , MAP Kinase Signaling System/drug effects , Markov Chains , Melanoma/drug therapy , Melanoma/pathology , NF-kappa B/metabolism , Phenotype , Proteome , Proteomics/methods , Proto-Oncogene Proteins B-raf/genetics
7.
Oncotarget ; 8(11): 17795-17809, 2017 Mar 14.
Article in English | MEDLINE | ID: mdl-28147313

ABSTRACT

Therapeutic resistance is a major obstacle to achieving durable clinical responses with targeted therapies, highlighting a need to elucidate the underlying mechanisms responsible for resistance and identify strategies to overcome this challenge. An emerging body of data implicates the tyrosine kinase MET in mediating resistance to BRAF inhibitors in BRAFV600E mutant melanoma. In this study we observed a dominant role for the HGF/MET axis in mediating resistance to BRAF and MEK inhibitors in models of BRAFV600E and NRAS mutant melanoma. In addition, we showed that MAPK pathway inhibition induced rapid increases in MET and GAB1 levels, providing novel mechanistic insight into how BRAFV600E mutant melanoma is primed for HGF-mediated rescue. We also determined that tumor-derived HGF, not systemic HGF, may be required to convey resistance to BRAF inhibition in vivo and that resistance could be reversed following treatment with AMG 337, a selective MET inhibitor. In summary, these findings support the clinical evaluation of MET-directed targeted therapy to circumvent resistance to BRAF and MEK inhibitors in BRAFV600E mutant melanoma. In addition, the induction of MET following treatment with BRAF and MEK inhibitors has the potential to serve as a predictive biomarker for identifying patients best suited for MET inhibitor combination therapy.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antineoplastic Agents/pharmacology , Hepatocyte Growth Factor/pharmacology , MAP Kinase Signaling System/drug effects , Melanoma/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Animals , Apoptosis/drug effects , Benzamides/pharmacology , Cell Line, Tumor , Dipeptides/pharmacology , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Drug Resistance, Neoplasm , Female , Humans , Indoles/pharmacology , Melanoma/genetics , Mice , Mice, Nude , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-met/metabolism , Pyridones/pharmacology , Sulfonamides/pharmacology , Triazoles/pharmacology , Vemurafenib , Xenograft Model Antitumor Assays
8.
Cancer Immunol Res ; 4(10): 845-857, 2016 10.
Article in English | MEDLINE | ID: mdl-27589875

ABSTRACT

The programmed cell death protein 1 (PD-1) limits effector T-cell functions in peripheral tissues, and its inhibition leads to clinical benefit in different cancers. To better understand how PD-1 blockade therapy modulates the tumor-host interactions, we evaluated three syngeneic murine tumor models, the BRAFV600E-driven YUMM1.1 and YUMM2.1 melanomas, and the carcinogen-induced murine colon adenocarcinoma MC38. The YUMM cell lines were established from mice with melanocyte-specific BRAFV600E mutation and PTEN loss (BRAFV600E/PTEN-/-). Anti-PD-1 or anti-PD-L1 therapy engendered strong antitumor activity against MC38 and YUMM2.1, but not YUMM1.1. PD-L1 expression did not differ between the three models at baseline or upon interferon stimulation. Whereas mutational load was high in MC38, it was lower in both YUMM models. In YUMM2.1, the antitumor activity of PD-1 blockade had a critical requirement for both CD4 and CD8 T cells, as well as CD28 and CD80/86 costimulation, with an increase in CD11c+CD11b+MHC-IIhigh dendritic cells and tumor-associated macrophages in the tumors after PD-1 blockade. Compared with YUMM1.1, YUMM2.1 exhibited a more inflammatory profile by RNA sequencing analysis, with an increase in expression of chemokine-trafficking genes that are related to immune cell recruitment and T-cell priming. In conclusion, response to PD-1 blockade therapy in tumor models requires CD4 and CD8 T cells and costimulation that is mediated by dendritic cells and macrophages. Cancer Immunol Res; 4(10); 845-57. ©2016 AACR.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Melanoma/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Dendritic Cells/immunology , Interferon-gamma/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Macrophages/immunology , Melanoma/genetics , Melanoma/immunology , Mice, Inbred C57BL , Mutation , Proto-Oncogene Proteins B-raf/genetics , Xenograft Model Antitumor Assays
9.
Semin Immunol ; 28(1): 73-80, 2016 02.
Article in English | MEDLINE | ID: mdl-26861544

ABSTRACT

Targeted therapies have induced high response rates and improved survival in patients with cancer. However, the long-term effectiveness of targeted therapies has been limited by the development of acquired resistance in the majority of patients. On the other hand, the modern immunotherapy strategies have been associated with durable responses but in limited number of patients. Accordingly, research efforts have been focused on examining the effects of combinations of targeted therapy and immunotherapy in several different histological subtypes of cancer. There has been accumulated evidence to suggest that targeted therapy can induce immune effects in the tumor cells, the host immune system, and the tumor microenvironment. Subsequently, clinical trials have been designed to examine the efficacy of combining immune checkpoint blockade or adoptive cell transfer with tyrosine kinase inhibitors, HER family blockade, anti-angiogenic agents, histone deacetylase inhibitors, and cancer stem cell inhibitors. To date, the combination of immunotherapy with targeted therapy has demonstrated potential as a cancer treatment strategy, but further optimizations are required and caution must be taken to avoid toxicity. The current review summarizes existing evidence and provides rationale supporting the use of combined targeted and immune-therapy approaches in patients with different types of cancer.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Cancer Vaccines/immunology , Immunotherapy, Adoptive/methods , Molecular Targeted Therapy , Neoplasms/therapy , Tumor Microenvironment/drug effects , Clinical Trials as Topic , Combined Modality Therapy , Histone Deacetylase Inhibitors/therapeutic use , Humans , Neoplasms/immunology , Neoplastic Stem Cells/physiology , Protein Kinase Inhibitors/therapeutic use , Receptor, ErbB-2/antagonists & inhibitors
11.
Semin Oncol ; 42(3): 466-73, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25965365

ABSTRACT

Immune-regulatory mechanisms are used by cancer to hide from the immune system. Advances and in-depth understanding of the biology of melanoma and its interaction with the immune system have led to the development of some of antagonistic antibodies to the programmed death 1 pathway (PD-1) and one of its ligands, programmed death ligand 1 (PD-L1), which are demonstrating high clinical benefit rates and tolerability. Blocking the immune-regulatory checkpoints that limit T-cell responses to melanoma upon PD-1/PD-L1 modulation has provided clinically validated targets for cancer immunotherapy. Combinations with other anti-melanoma agents may result in additional benefits. Nivolumab, pembrolizumab (formerly known as MK-3475 and lambrolizumab), and pidilizumab are anti-PD-1 antibodies in clinical development for melanoma, non-small cell lung cancer, renal cell carcinoma, head and neck cancers, lymphoma, and several other cancers. Long-term survivors already have been reported with these therapies. In this review, we discuss the current state of anti-PD-1 agents, the evidence in the literature to support the combination of anti-PD-1 antibodies with other anti-cancer agents and discuss the future directions for rational design of clinical trials that keep on increasing the number of long-term survivors.


Subject(s)
Antineoplastic Agents/therapeutic use , Melanoma/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Humans , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Immunotherapy , Melanoma/immunology , Melanoma/metabolism , Molecular Targeted Therapy , Programmed Cell Death 1 Receptor/metabolism
12.
Sci Transl Med ; 7(279): 279ra41, 2015 Mar 18.
Article in English | MEDLINE | ID: mdl-25787767

ABSTRACT

Combining immunotherapy and BRAF targeted therapy may result in improved antitumor activity with the high response rates of targeted therapy and the durability of responses with immunotherapy. However, the first clinical trial testing the combination of the BRAF inhibitor vemurafenib and the CTLA4 antibody ipilimumab was terminated early because of substantial liver toxicities. MEK [MAPK (mitogen-activated protein kinase) kinase] inhibitors can potentiate the MAPK inhibition in BRAF mutant cells while potentially alleviating the unwanted paradoxical MAPK activation in BRAF wild-type cells that lead to side effects when using BRAF inhibitors alone. However, there is the concern of MEK inhibitors being detrimental to T cell functionality. Using a mouse model of syngeneic BRAF(V600E)-driven melanoma, SM1, we tested whether addition of the MEK inhibitor trametinib would enhance the antitumor activity of combined immunotherapy with the BRAF inhibitor dabrafenib. Combination of dabrafenib and trametinib with pmel-1 adoptive cell transfer (ACT) showed complete tumor regression, increased T cell infiltration into tumors, and improved in vivo cytotoxicity. Single-agent dabrafenib increased tumor-associated macrophages and T regulatory cells (Tregs) in tumors, which decreased with the addition of trametinib. The triple combination therapy resulted in increased melanosomal antigen and major histocompatibility complex (MHC) expression and global immune-related gene up-regulation. Given the up-regulation of PD-L1 seen with dabrafenib and/or trametinib combined with antigen-specific ACT, we tested the combination of dabrafenib, trametinib, and anti-PD1 therapy in SM1 tumors, and observed superior antitumor effect. Our findings support the testing of triple combination therapy of BRAF and MEK inhibitors with immunotherapy in patients with BRAF(V600E) mutant metastatic melanoma.


Subject(s)
Antineoplastic Agents/therapeutic use , Immunotherapy/methods , MAP Kinase Kinase Kinases/antagonists & inhibitors , Melanoma/drug therapy , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Skin Neoplasms/drug therapy , Animals , Antigens, Neoplasm/metabolism , Antineoplastic Agents/chemistry , CTLA-4 Antigen/immunology , Cell Line, Tumor , Cell Survival , Humans , Imidazoles/chemistry , MAP Kinase Kinase Kinases/immunology , Major Histocompatibility Complex , Melanoma/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oximes/chemistry , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/immunology , Pyridones/chemistry , Pyrimidinones/chemistry , Skin Neoplasms/immunology
13.
Proc Natl Acad Sci U S A ; 112(5): E420-9, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25605940

ABSTRACT

Canonical Wnt signaling plays an important role in development and disease, regulating transcription of target genes and stabilizing many proteins phosphorylated by glycogen synthase kinase 3 (GSK3). We observed that the MiT family of transcription factors, which includes the melanoma oncogene MITF (micropthalmia-associated transcription factor) and the lysosomal master regulator TFEB, had the highest phylogenetic conservation of three consecutive putative GSK3 phosphorylation sites in animal proteomes. This finding prompted us to examine the relationship between MITF, endolysosomal biogenesis, and Wnt signaling. Here we report that MITF expression levels correlated with the expression of a large subset of lysosomal genes in melanoma cell lines. MITF expression in the tetracycline-inducible C32 melanoma model caused a marked increase in vesicular structures, and increased expression of late endosomal proteins, such as Rab7, LAMP1, and CD63. These late endosomes were not functional lysosomes as they were less active in proteolysis, yet were able to concentrate Axin1, phospho-LRP6, phospho-ß-catenin, and GSK3 in the presence of Wnt ligands. This relocalization significantly enhanced Wnt signaling by increasing the number of multivesicular bodies into which the Wnt signalosome/destruction complex becomes localized upon Wnt signaling. We also show that the MITF protein was stabilized by Wnt signaling, through the novel C-terminal GSK3 phosphorylations identified here. MITF stabilization caused an increase in multivesicular body biosynthesis, which in turn increased Wnt signaling, generating a positive-feedback loop that may function during the proliferative stages of melanoma. The results underscore the importance of misregulated endolysosomal biogenesis in Wnt signaling and cancer.


Subject(s)
Endosomes/physiology , Lysosomes/physiology , Microphthalmia-Associated Transcription Factor/physiology , Signal Transduction/physiology , Wnt Proteins/metabolism , Cell Line, Tumor , Glycogen Synthase Kinase 3/metabolism , HEK293 Cells , Humans , Phosphorylation
14.
Nat Commun ; 5: 5712, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25502142

ABSTRACT

Increased expression of the Microphthalmia-associated transcription factor (MITF) contributes to melanoma progression and resistance to BRAF pathway inhibition. Here we show that the lack of MITF is associated with more severe resistance to a range of inhibitors, while its presence is required for robust drug responses. Both in primary and acquired resistance, MITF levels inversely correlate with the expression of several activated receptor tyrosine kinases, most frequently AXL. The MITF-low/AXL-high/drug-resistance phenotype is common among mutant BRAF and NRAS melanoma cell lines. The dichotomous behaviour of MITF in drug response is corroborated in vemurafenib-resistant biopsies, including MITF-high and -low clones in a relapsed patient. Furthermore, drug cocktails containing AXL inhibitor enhance melanoma cell elimination by BRAF or ERK inhibition. Our results demonstrate that a low MITF/AXL ratio predicts early resistance to multiple targeted drugs, and warrant clinical validation of AXL inhibitors to combat resistance of BRAF and NRAS mutant MITF-low melanomas.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Melanoma/drug therapy , Microphthalmia-Associated Transcription Factor/genetics , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Skin Neoplasms/drug therapy , Aminopyridines/pharmacology , Animals , Benzamides/pharmacology , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Imatinib Mesylate , Imidazoles/pharmacology , Indoles/pharmacology , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Mice , Microphthalmia-Associated Transcription Factor/metabolism , Oximes/pharmacology , Piperazines/pharmacology , Prognosis , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Pyridones/pharmacology , Pyrimidines/pharmacology , Pyrimidinones/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Sulfonamides/pharmacology , Vemurafenib , Xenograft Model Antitumor Assays , Axl Receptor Tyrosine Kinase
15.
Nature ; 515(7528): 568-71, 2014 Nov 27.
Article in English | MEDLINE | ID: mdl-25428505

ABSTRACT

Therapies that target the programmed death-1 (PD-1) receptor have shown unprecedented rates of durable clinical responses in patients with various cancer types. One mechanism by which cancer tissues limit the host immune response is via upregulation of PD-1 ligand (PD-L1) and its ligation to PD-1 on antigen-specific CD8(+) T cells (termed adaptive immune resistance). Here we show that pre-existing CD8(+) T cells distinctly located at the invasive tumour margin are associated with expression of the PD-1/PD-L1 immune inhibitory axis and may predict response to therapy. We analysed samples from 46 patients with metastatic melanoma obtained before and during anti-PD-1 therapy (pembrolizumab) using quantitative immunohistochemistry, quantitative multiplex immunofluorescence, and next-generation sequencing for T-cell antigen receptors (TCRs). In serially sampled tumours, patients responding to treatment showed proliferation of intratumoral CD8(+) T cells that directly correlated with radiographic reduction in tumour size. Pre-treatment samples obtained from responding patients showed higher numbers of CD8-, PD-1- and PD-L1-expressing cells at the invasive tumour margin and inside tumours, with close proximity between PD-1 and PD-L1, and a more clonal TCR repertoire. Using multivariate analysis, we established a predictive model based on CD8 expression at the invasive margin and validated the model in an independent cohort of 15 patients. Our findings indicate that tumour regression after therapeutic PD-1 blockade requires pre-existing CD8(+) T cells that are negatively regulated by PD-1/PD-L1-mediated adaptive immune resistance.


Subject(s)
Adaptive Immunity/immunology , CD8-Positive T-Lymphocytes/immunology , Immunotherapy , Melanoma/therapy , Models, Biological , Aged , Aged, 80 and over , Biomarkers , CD8-Positive T-Lymphocytes/cytology , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Melanoma/diagnosis , Melanoma/immunology , Melanoma/pathology , Middle Aged , Multivariate Analysis , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Treatment Outcome
16.
Oncoimmunology ; 3: e29244, 2014.
Article in English | MEDLINE | ID: mdl-25083336

ABSTRACT

Targeting immune inhibitory receptors has brought excitement, innovation and hope to cancer patients. Our recent work revealed the immunological effects of blocking the CTLA4 and PD-1 immune checkpoints on T cell receptor usage among peripheral blood cells, and further uncovers how the expansion of the T cell repertoire matches the immunotoxicity profile of the therapy.

17.
Mol Cancer ; 13: 194, 2014 Aug 20.
Article in English | MEDLINE | ID: mdl-25142146

ABSTRACT

BACKGROUND: In melanoma, dysregulation of the MAPK pathway, usually via BRAF(V600) or NRAS(Q61) somatic mutations, leads to constitutive ERK signaling. While BRAF inhibitors are initially effective for BRAF-mutant melanoma, no FDA-approved targeted therapies exist for BRAF-inhibitor-resistant BRAF(V600), NRAS mutant, or wild-type melanoma. METHODS: The 50% inhibitory concentration (IC50) of SCH772984, a novel inhibitor of ERK1/2, was determined in a panel of 50 melanoma cell lines. Effects on MAPK and AKT signaling by western blotting and cell cycle by flow cytometry were determined. RESULTS: Sensitivity fell into three groups: sensitive, 50% inhibitory concentration (IC50) < 1 µM; intermediately sensitive, IC50 1-2 µM; and resistant, >2 µM. Fifteen of 21 (71%) BRAF mutants, including 4 with innate vemurafenib resistance, were sensitive to SCH772984. All three (100%) BRAF/NRAS double mutants, 11 of 14 (78%) NRAS mutants and 5 of 7 (71%) wild-type melanomas were sensitive. Among BRAF(V600) mutants with in vitro acquired resistance to vemurafenib, those with MAPK pathway reactivation as the mechanism of resistance were sensitive to SCH772984. SCH772984 caused G1 arrest and induced apoptosis. CONCLUSIONS: Combining vemurafenib and SCH722984 in BRAF mutant melanoma was synergistic in a majority of cell lines and significantly delayed the onset of acquired resistance in long term in vitro assays. Therefore, SCH772984 may be clinically applicable as a treatment for non-BRAF mutant melanoma or in BRAF-mutant melanoma with innate or acquired resistance, alone or in combination with BRAF inhibitors.


Subject(s)
GTP Phosphohydrolases/antagonists & inhibitors , Indazoles/pharmacology , Membrane Proteins/antagonists & inhibitors , Multiple Myeloma/pathology , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Synergism , GTP Phosphohydrolases/genetics , Humans , Indoles/pharmacology , Inhibitory Concentration 50 , Membrane Proteins/genetics , Molecular Targeted Therapy , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Mutation , Proto-Oncogene Proteins B-raf/genetics , Sulfonamides/pharmacology , Vemurafenib
18.
J Clin Oncol ; 32(21): 2248-54, 2014 Jul 20.
Article in English | MEDLINE | ID: mdl-24958825

ABSTRACT

Recent breakthroughs in the treatment of advanced melanoma are based on scientific advances in understanding oncogenic signaling and the immunobiology of this cancer. Targeted therapy can successfully block oncogenic signaling in BRAF(V600)-mutant melanoma with high initial clinical responses, but relapse rates are also high. Activation of an immune response by releasing inhibitory check points can induce durable responses in a subset of patients with melanoma. These advances have driven interest in combining both modes of therapy with the goal of achieving high response rates with prolonged duration. Combining BRAF inhibitors and immunotherapy can specifically target the BRAF(V600) driver mutation in the tumor cells and potentially sensitize the immune system to target tumors. However, it is becoming evident that the effects of paradoxical mitogen-activated protein kinase pathway activation by BRAF inhibitors in non-BRAF-mutant cells needs to be taken into account, which may be implicated in the problems encountered in the first clinical trial testing a combination of the BRAF inhibitor vemurafenib with ipilimumab (anti-CTLA4), with significant liver toxicities. Here, we present the concept and potential mechanisms of combinatorial activity of targeted therapy and immunotherapy, review the literature for evidence to support the combination, and discuss the potential challenges and future directions for rational conduct of clinical trials.


Subject(s)
Immunotherapy , Melanoma/therapy , Molecular Targeted Therapy , Mutation/genetics , Proto-Oncogene Proteins B-raf/genetics , Antineoplastic Combined Chemotherapy Protocols , Humans , Melanoma/genetics , Prognosis , Protein Kinase Inhibitors/therapeutic use
19.
Clin Cancer Res ; 20(13): 3446-57, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24812408

ABSTRACT

PURPOSE: PD-L1 is the main ligand for the immune inhibitory receptor PD-1. This ligand is frequently expressed by melanoma cells. In this study, we investigated whether PD-L1 expression is controlled by melanoma driver mutations and modified by oncogenic signaling inhibition. EXPERIMENTAL DESIGN: Expression of PD-L1 was investigated in a panel of 51 melanoma cell lines containing different oncogenic mutations, including cell lines with innate and acquired resistance to BRAF inhibitors (BRAFi). The effects of targeted therapy drugs on expression of PD-L1 by melanoma cells were investigated. RESULTS: No association was found between the level of PD-L1 expression and mutations in BRAF, NRAS, PTEN, or amplification of AKT. Resistance to vemurafenib due to the activation of alternative signaling pathways was accompanied with the induction of PD-L1 expression, whereas the resistance due to the reactivation of the MAPK pathway had no effect on PD-L1 expression. In melanoma cell lines, the effects of BRAF, MEK, and PI3K inhibitors on expression of PD-L1 were variable from reduction to induction, particularly in the presence of INFγ. In PD-L1-exposed lymphocytes, vemurafenib paradoxically restored activity of the MAPK pathway and increased the secretion of cytokines. CONCLUSIONS: In melanoma cell lines, including BRAFi-resistant cells, PD-L1 expression is variably regulated by oncogenic signaling pathways. PD-L1-exposed lymphocytes decrease MAPK signaling, which is corrected by exposure to vemurafenib, providing potential benefits of combining this drug with immunotherapies.


Subject(s)
B7-H1 Antigen/genetics , Melanoma/genetics , Melanoma/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Signal Transduction , B7-H1 Antigen/metabolism , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Cytokines/biosynthesis , Drug Resistance, Neoplasm/genetics , Humans , Indoles/pharmacology , Lymphocytes/immunology , Lymphocytes/metabolism , Melanoma/immunology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mutation , Signal Transduction/drug effects , Sulfonamides/pharmacology , Vemurafenib
20.
Mol Cancer ; 13: 83, 2014 Apr 16.
Article in English | MEDLINE | ID: mdl-24735930

ABSTRACT

BACKGROUND: The clinical use of BRAF inhibitors for treatment of metastatic melanoma is limited by the development of drug resistance. In this study we investigated whether co-targeting the MAPK and the PI3K-AKT pathway can prevent emergence of resistance or provide additional growth inhibitory effects in vitro. METHODS: Anti-tumor effects of the combination of the BRAF inhibitor (BRAFi) dabrafenib and GSK2141795B (AKTi) in a panel of 23 BRAF mutated melanoma cell lines were evaluated on growth inhibition by an ATP-based luminescent assay, on cell cycle and apoptosis by flow cytometry and on cell signaling by western blot. Moreover, we investigated the possibilities of delaying or reversing resistance or achieving further growth inhibition by combining AKTi with dabrafenib and/or the MEK inhibitor (MEKi) trametinib by using long term cultures. RESULTS: More than 40% of the cell lines, including PTEN-/- and AKT mutants showed sensitivity to AKTi (IC50 < 1.5 µM). The combination of dabrafenib and AKTi synergistically potentiated growth inhibition in the majority of cell lines with IC50 > 5 nM dabrafenib. Combinatorial treatment induced apoptosis only in cell lines sensitive to AKTi. In long term cultures of a PTEN-/- cell line, combinatorial treatment with the MAPK inhibitors, dabrafenib and trametinib, and AKTi markedly delayed the emergence of drug resistance. Moreover, combining AKTi with the MAPK inhibitors from the beginning provided superior growth inhibitory effects compared to addition of AKTi upon development of resistance to MAPK inhibitors in this particular cell line. CONCLUSIONS: AKTi combined with BRAFi-based therapy may benefit patients with tumors harboring BRAF mutations and particularly PTEN deletions or AKT mutations.


Subject(s)
Drug Resistance, Neoplasm/genetics , Melanoma/drug therapy , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-akt/genetics , Apoptosis/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Humans , Imidazoles/administration & dosage , MAP Kinase Kinase Kinases/antagonists & inhibitors , Melanoma/genetics , Melanoma/pathology , Mutation , Oximes/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyridones/administration & dosage , Pyrimidinones/administration & dosage , Signal Transduction , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...