Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
bioRxiv ; 2023 Nov 14.
Article in English | MEDLINE | ID: mdl-38014031

ABSTRACT

Microphthalmia-associated transcription factor (MITF) plays pivotal roles in melanocyte development, function, and melanoma pathogenesis. MITF amplification occurs in melanoma and has been associated with resistance to targeted therapies. Here, we show that MITF regulates a global antioxidant program that increases survival of melanoma cell lines by protecting the cells from reactive oxygen species (ROS)-induced damage. In addition, this redox program is correlated with MITF expression in human melanoma cell lines and patient-derived melanoma samples. Using a zebrafish melanoma model, we show that MITF decreases ROS-mediated DNA damage in vivo . Some of the MITF target genes involved, such as IDH1 and NNT , are regulated through direct MITF binding to canonical enhancer box (E-BOX) sequences proximal to their promoters. Utilizing functional experiments, we demonstrate the role of MITF and its target genes in reducing cytosolic and mitochondrial ROS. Collectively, our data identify MITF as a significant driver of the cellular antioxidant state. One Sentence Summary: MITF promote melanoma survival via increasing ROS tolerance.

2.
Front Immunol ; 13: 833636, 2022.
Article in English | MEDLINE | ID: mdl-35185925

ABSTRACT

The establishment of an "interferon (IFN) signature" to subset SLE patients on disease severity has led to therapeutics targeting IFNα. Here, we investigate IFN signaling in SLE using multiplexed protein arrays and single cell cytometry by time of flight (CyTOF). First, the IFN signature for SLE patients (n=81) from the Stanford Lupus Registry is determined using fluidigm qPCR measuring 44 previously determined IFN-inducible transcripts. IFN-high (IFN-H) patients have increased SLE criteria and renal/CNS/immunologic involvement, and increased autoantibody reactivity against spliceosome-associated antigens. CyTOF analysis is performed on non-stimulated and stimulated (IFNα, IFNγ, IL-21) PBMCs from SLE patients (n=25) and HCs (n=9) in a panel identifying changes in phosphorylation of intracellular signaling proteins (pTOF). Another panel is utilized to detect changes in intracellular cytokine (ICTOF) production in non-stimulated and stimulated (PMA/ionomycin) PBMCs from SLE patients (n=31) and HCs (n=17). Bioinformatic analysis by MetaCyto and OMIQ reveal phenotypic changes in immune cell subsets between IFN-H and IFN-low (IFN-L) patients. Most notably, IFN-H patients exhibit increased STAT1/3/5 phosphorylation downstream of cytokine stimulation and increased phosphorylation of non-canonical STAT proteins. These results suggest that IFN signaling in SLE modulates STAT phosphorylation, potentially uncovering possible targets for future therapeutic approaches.


Subject(s)
Interferon Type I/physiology , Interleukins/physiology , Leukocytes, Mononuclear/metabolism , Lupus Erythematosus, Systemic/metabolism , STAT1 Transcription Factor/metabolism , Adult , Female , Flow Cytometry , Humans , Interferon Type I/analysis , Interleukins/analysis , Male , Middle Aged , Phosphorylation , Signal Transduction , Single-Cell Analysis
3.
J Clin Invest ; 131(12)2021 06 15.
Article in English | MEDLINE | ID: mdl-33914706

ABSTRACT

Melanoma dedifferentiation has been reported to be a state of cellular resistance to targeted therapies and immunotherapies as cancer cells revert to a more primitive cellular phenotype. Here, we show that, counterintuitively, the biopsies of patient tumors that responded to anti-programmed cell death 1 (anti-PD-1) therapy had decreased expression of melanocytic markers and increased neural crest markers, suggesting treatment-induced dedifferentiation. When modeling the effects in vitro, we documented that melanoma cell lines that were originally differentiated underwent a process of neural crest dedifferentiation when continuously exposed to IFN-γ, through global chromatin landscape changes that led to enrichment in specific hyperaccessible chromatin regions. The IFN-γ-induced dedifferentiation signature corresponded with improved outcomes in patients with melanoma, challenging the notion that neural crest dedifferentiation is entirely an adverse phenotype.


Subject(s)
Biomarkers, Tumor , Cell Dedifferentiation/drug effects , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Immune Checkpoint Inhibitors/pharmacology , Interferon-gamma/metabolism , Melanoma , Neoplasm Proteins , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Humans , Melanocytes/metabolism , Melanocytes/pathology , Melanoma/drug therapy , Melanoma/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism
5.
Science ; 370(6520): 1099-1104, 2020 11 27.
Article in English | MEDLINE | ID: mdl-33243890

ABSTRACT

Living cells often identify their correct partner or target cells by integrating information from multiple receptors, achieving levels of recognition that are difficult to obtain with individual molecular interactions. In this study, we engineered a diverse library of multireceptor cell-cell recognition circuits by using synthetic Notch receptors to transcriptionally interconnect multiple molecular recognition events. These synthetic circuits allow engineered T cells to integrate extra- and intracellular antigen recognition, are robust to heterogeneity, and achieve precise recognition by integrating up to three different antigens with positive or negative logic. A three-antigen AND gate composed of three sequentially linked receptors shows selectivity in vivo, clearing three-antigen tumors while ignoring related two-antigen tumors. Daisy-chaining multiple molecular recognition events together in synthetic circuits provides a powerful way to engineer cellular-level recognition.


Subject(s)
Cell Communication/immunology , Cell Engineering , Receptors, Chimeric Antigen/immunology , Receptors, Notch/immunology , T-Lymphocytes/immunology , Animals , Antigens, Neoplasm/immunology , Humans , Mice , Receptors, Chimeric Antigen/genetics , Receptors, Notch/genetics , Transcription, Genetic
6.
Cancer Cell ; 38(4): 500-515.e3, 2020 10 12.
Article in English | MEDLINE | ID: mdl-32916126

ABSTRACT

We analyze the transcriptome of baseline and on-therapy tumor biopsies from 101 patients with advanced melanoma treated with nivolumab (anti-PD-1) alone or combined with ipilimumab (anti-CTLA-4). We find that T cell infiltration and interferon-γ (IFN-γ) signaling signatures correspond most highly with clinical response to therapy, with a reciprocal decrease in cell-cycle and WNT signaling pathways in responding biopsies. We model the interaction in 58 human cell lines, where IFN-γ in vitro exposure leads to a conserved transcriptome response unless cells have IFN-γ receptor alterations. This conserved IFN-γ transcriptome response in melanoma cells serves to amplify the antitumor immune response. Therefore, the magnitude of the antitumor T cell response and the corresponding downstream IFN-γ signaling are the main drivers of clinical response or resistance to immune checkpoint blockade therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Interferon-gamma/metabolism , Melanoma/drug therapy , Adult , Aged , Aged, 80 and over , Cell Line , Cell Line, Tumor , Female , Gene Expression Profiling/methods , Humans , Immune Checkpoint Inhibitors/administration & dosage , Interferon-gamma/pharmacology , Ipilimumab/administration & dosage , Male , Melanoma/genetics , Melanoma/metabolism , Middle Aged , Nivolumab/administration & dosage , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Transcriptome/drug effects , Transcriptome/genetics , Young Adult
7.
Cancer Discov ; 10(8): 1140-1157, 2020 08.
Article in English | MEDLINE | ID: mdl-32467343

ABSTRACT

Mechanism-based strategies to overcome resistance to PD-1 blockade therapy are urgently needed. We developed genetic acquired resistant models of JAK1, JAK2, and B2M loss-of-function mutations by gene knockout in human and murine cell lines. Human melanoma cell lines with JAK1/2 knockout became insensitive to IFN-induced antitumor effects, while B2M knockout was no longer recognized by antigen-specific T cells and hence was resistant to cytotoxicity. All of these mutations led to resistance to anti-PD-1 therapy in vivo. JAK1/2-knockout resistance could be overcome with the activation of innate and adaptive immunity by intratumoral Toll-like receptor 9 agonist administration together with anti-PD-1, mediated by natural killer (NK) and CD8 T cells. B2M-knockout resistance could be overcome by NK-cell and CD4 T-cell activation using the CD122 preferential IL2 agonist bempegaldesleukin. Therefore, mechanistically designed combination therapies can overcome genetic resistance to PD-1 blockade therapy. SIGNIFICANCE: The activation of IFN signaling through pattern recognition receptors and the stimulation of NK cells overcome genetic mechanisms of resistance to PD-1 blockade therapy mediated through deficient IFN receptor and antigen presentation pathways. These approaches are being tested in the clinic to improve the antitumor activity of PD-1 blockade therapy.This article is highlighted in the In This Issue feature, p. 1079.


Subject(s)
Drug Resistance, Neoplasm/genetics , Janus Kinase 1/genetics , Janus Kinase 2/genetics , Programmed Cell Death 1 Receptor/antagonists & inhibitors , beta 2-Microglobulin/genetics , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Interferons/pharmacology , Interleukin-2/analogs & derivatives , Interleukin-2/immunology , Interleukin-2/pharmacology , Interleukin-2/therapeutic use , Killer Cells, Natural/immunology , Loss of Function Mutation , Mice, Inbred C57BL , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/immunology , Polyethylene Glycols/pharmacology , Polyethylene Glycols/therapeutic use , Toll-Like Receptor 9/immunology
8.
Nat Commun ; 11(1): 660, 2020 01 31.
Article in English | MEDLINE | ID: mdl-32005809

ABSTRACT

Interleukin-2 (IL-2) is a component of most protocols of adoptive cell transfer (ACT) therapy for cancer, but is limited by short exposure and high toxicities. NKTR-214 is a kinetically-engineered IL-2 receptor ßγ (IL-2Rßγ)-biased agonist consisting of IL-2 conjugated to multiple releasable polyethylene glycol chains resulting in sustained signaling through IL-2Rßγ. We report that ACT supported by NKTR-214 increases the proliferation, homing and persistence of anti-tumor T cells compared to ACT with IL-2, resulting in superior antitumor activity in a B16-F10 murine melanoma model. The use of NKTR-214 increases the number of polyfunctional T cells in murine spleens and tumors compared to IL-2, and enhances the polyfunctionality of T and NK cells in the peripheral blood of patients receiving NKTR-214 in a phase 1 trial. In conclusion, NKTR-214 may have the potential to improve the antitumor activity of ACT in humans through increased in vivo expansion and polyfunctionality of the adoptively transferred T cells.


Subject(s)
Adoptive Transfer , Interleukin-2/analogs & derivatives , Interleukin-2/agonists , Melanoma/drug therapy , Polyethylene Glycols/administration & dosage , Receptors, Interleukin-2/immunology , T-Lymphocytes/immunology , Animals , Humans , Interleukin-2/administration & dosage , Interleukin-2/immunology , Lymphocyte Activation/drug effects , Melanoma/genetics , Melanoma/immunology , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Receptors, Interleukin-2/genetics
10.
Nat Cancer ; 1(1): 46-58, 2020.
Article in English | MEDLINE | ID: mdl-34368780

ABSTRACT

Lack of tumor infiltration by immune cells is the main mechanism of primary resistance to programmed cell death protein 1 (PD-1) blockade therapies for cancer. It has been postulated that cancer cell-intrinsic mechanisms may actively exclude T cells from tumors, suggesting that the finding of actionable molecules that could be inhibited to increase T cell infiltration may synergize with checkpoint inhibitor immunotherapy. Here, we show that p21-activated kinase 4 (PAK4) is enriched in non-responding tumor biopsies with low T cell and dendritic cell infiltration. In mouse models, genetic deletion of PAK4 increased T cell infiltration and reversed resistance to PD-1 blockade in a CD8 T cell-dependent manner. Furthermore, combination of anti-PD-1 with the PAK4 inhibitor KPT-9274 improved anti-tumor response compared with anti-PD-1 alone. Therefore, high PAK4 expression is correlated with low T cell and dendritic cell infiltration and a lack of response to PD-1 blockade, which could be reversed with PAK4 inhibition.


Subject(s)
Immune Checkpoint Inhibitors , Immunotherapy , Neoplasms , Programmed Cell Death 1 Receptor , p21-Activated Kinases , Animals , CD8-Positive T-Lymphocytes , Mice , Neoplasms/drug therapy , p21-Activated Kinases/genetics
12.
Nat Med ; 25(6): 936-940, 2019 06.
Article in English | MEDLINE | ID: mdl-31171879

ABSTRACT

Oncogene-targeted therapy with B-Raf proto-oncogene (BRAF) and mitogen-activated protein kinase kinase (MEK) inhibitors induces a high initial response rate in patients with BRAFV600-mutated melanoma, with a median duration of response of approximately 1 year1-3. Immunotherapy with antibodies to programmed death 1 (PD-1) produces lower response rates but with long response duration. Preclinical models suggest that combining BRAF and MEK inhibitors with PD-1 blockade therapy improves antitumor activity4-6, which may provide additional treatment options for patients unlikely to have long-lasting responses to either mode of therapy alone. We enrolled 15 patients with BRAFV600-mutated metastatic melanoma in a first-in-human clinical trial of dabrafenib, trametinib and pembrolizumab ( NCT02130466 ). Eleven patients (73%) experienced grade 3/4 treatment-related adverse events, the most common being elevation of liver function tests and pyrexia, most of which resolved with drug interruption or discontinuation of either the anti-PD-1 antibody or the targeted therapy combination. Eleven patients (73%; 95% confidence interval = 45-92%) had an objective response, and six (40%; 95% confidence interval = 16-68%) continued with a response at a median follow-up of 27 months (range = 10.3-38.4+ months) for all patients. This study suggests that this triple-combined therapy may benefit a subset of patients with BRAFV600-mutated metastatic melanoma by increasing the frequency of long-lasting antitumor responses.


Subject(s)
MAP Kinase Kinase Kinases/antagonists & inhibitors , Melanoma/drug therapy , Melanoma/genetics , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Adult , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Female , Humans , Imidazoles/administration & dosage , Immunotherapy , Male , Melanoma/secondary , Middle Aged , Mutation , Oximes/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Mas , Pyridones/administration & dosage , Pyrimidinones/administration & dosage , Skin Neoplasms/therapy , Young Adult
13.
J Transl Med ; 17(1): 113, 2019 04 05.
Article in English | MEDLINE | ID: mdl-30953519

ABSTRACT

BACKGROUND: Various proinflammatory cytokines can be detected within the melanoma tumor microenvironment. Interleukin 32 (IL32) is produced by T cells, NK cells and monocytes/macrophages, but also by a subset of melanoma cells. We sought to better understand the biology of IL32 in human melanoma. METHODS: We analyzed RNA sequencing data from 53 in-house established human melanoma cell lines and 479 melanoma tumors from The Cancer Genome Atlas dataset. We evaluated global gene expression patterns associated with IL32 expression. We also evaluated the impact of proinflammatory molecules TNFα and IFNγ on IL32 expression and dedifferentiation in melanoma cell lines in vitro. In order to study the transcriptional regulation of IL32 in these cell lines, we cloned up to 10.5 kb of the 5' upstream region of the human IL32 gene into a luciferase reporter vector. RESULTS: A significant proportion of established human melanoma cell lines express IL32, with its expression being highly correlated with a dedifferentiation genetic signature (high AXL/low MITF). Non IL32-expressing differentiated melanoma cell lines exposed to TNFα or IFNγ can be induced to express the three predominant isoforms (α, ß, γ) of IL32. Cis-acting elements within this 5' upstream region of the human IL32 gene appear to govern both induced and constitutive gene expression. In the tumor microenvironment, IL32 expression is highly correlated with genes related to T cell infiltration, and also positively correlates with high AXL/low MITF dedifferentiated gene signature. CONCLUSIONS: Expression of IL32 in human melanoma can be induced by TNFα or IFNγ and correlates with a treatment-resistant dedifferentiated genetic signature. Constitutive and induced expression are regulated, in part, by cis-acting sequences within the 5' upstream region.


Subject(s)
Interleukins/genetics , Melanoma/genetics , Skin Neoplasms/genetics , Biopsy , Cell Dedifferentiation/genetics , Cell Line, Tumor , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Interferon-gamma/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Melanoma/pathology , Microarray Analysis , Neoplasm Metastasis , Skin Neoplasms/pathology , Transcriptome , Tumor Microenvironment/genetics , Tumor Necrosis Factor-alpha/metabolism
14.
Clin Cancer Res ; 25(7): 2096-2108, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30573690

ABSTRACT

PURPOSE: Transgenic adoptive cell therapy (ACT) targeting the tumor antigen NY-ESO-1 can be effective for the treatment of sarcoma and melanoma. Preclinical models have shown that this therapy can be improved with the addition of dendritic cell (DC) vaccination and immune checkpoint blockade. We studied the safety, feasibility, and antitumor efficacy of transgenic ACT with DC vaccination, with and without CTLA-4 blockade with ipilimumab. PATIENTS AND METHODS: Freshly prepared autologous NY-ESO-1-specific T-cell receptor (TCR) transgenic lymphocytes were adoptively transferred together with NY-ESO-1 peptide-pulsed DC vaccination in HLA-A2.1-positive subjects alone (ESO, NCT02070406) or with ipilimumab (INY, NCT01697527) in patients with advanced sarcoma or melanoma. RESULTS: Six patients were enrolled in the ESO cohort, and four were enrolled in the INY cohort. Four out of six patients treated per ESO (66%), and two out of four patients treated per INY (50%) displayed evidence of tumor regression. Peripheral blood reconstitution with NY-ESO-1-specific T cells peaked within 2 weeks of ACT, indicating rapid in vivo expansion. Tracking of transgenic T cells to the tumor sites was demonstrated in on-treatment biopsies via TCR sequencing. Multiparametric mass cytometry of transgenic cells demonstrated shifting of transgenic cells from memory phenotypes to more terminally differentiated effector phenotypes over time. CONCLUSIONS: ACT of fresh NY-ESO-1 transgenic T cells prepared via a short ex vivo protocol and given with DC vaccination, with or without ipilimumab, is feasible and results in transient antitumor activity, with no apparent clinical benefit of the addition of ipilimumab. Improvements are needed to maintain tumor responses.


Subject(s)
Adoptive Transfer , Antineoplastic Agents, Immunological/pharmacology , Cancer Vaccines/immunology , Dendritic Cells/immunology , Ipilimumab/pharmacology , Neoplasms/immunology , Neoplasms/therapy , Adoptive Transfer/methods , Adult , Animals , CTLA-4 Antigen/antagonists & inhibitors , Cell Line, Tumor , Combined Modality Therapy , Dendritic Cells/metabolism , Female , Gene Knock-In Techniques , Humans , Immunotherapy , Lymphocytes/immunology , Lymphocytes/metabolism , Male , Mice , Middle Aged , Molecular Targeted Therapy , Neoplasms/pathology , Phenotype , Pilot Projects , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Young Adult
15.
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
16.
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
17.
Cancer Discov ; 8(6): 730-749, 2018 06.
Article in English | MEDLINE | ID: mdl-29510987

ABSTRACT

To understand the genetic drivers of immune recognition and evasion in colorectal cancer, we analyzed 1,211 colorectal cancer primary tumor samples, including 179 classified as microsatellite instability-high (MSI-high). This set includes The Cancer Genome Atlas colorectal cancer cohort of 592 samples, completed and analyzed here. MSI-high, a hypermutated, immunogenic subtype of colorectal cancer, had a high rate of significantly mutated genes in important immune-modulating pathways and in the antigen presentation machinery, including biallelic losses of B2M and HLA genes due to copy-number alterations and copy-neutral loss of heterozygosity. WNT/ß-catenin signaling genes were significantly mutated in all colorectal cancer subtypes, and activated WNT/ß-catenin signaling was correlated with the absence of T-cell infiltration. This large-scale genomic analysis of colorectal cancer demonstrates that MSI-high cases frequently undergo an immunoediting process that provides them with genetic events allowing immune escape despite high mutational load and frequent lymphocytic infiltration and, furthermore, that colorectal cancer tumors have genetic and methylation events associated with activated WNT signaling and T-cell exclusion.Significance: This multi-omic analysis of 1,211 colorectal cancer primary tumors reveals that it should be possible to better monitor resistance in the 15% of cases that respond to immune blockade therapy and also to use WNT signaling inhibitors to reverse immune exclusion in the 85% of cases that currently do not. Cancer Discov; 8(6); 730-49. ©2018 AACR.This article is highlighted in the In This Issue feature, p. 663.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Tumor Escape , DNA Copy Number Variations , DNA Methylation , Germ-Line Mutation , HLA Antigens/genetics , Humans , Loss of Heterozygosity , Microsatellite Instability , Wnt Signaling Pathway , beta 2-Microglobulin/genetics
18.
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
19.
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
20.
Sci Rep ; 6: 27454, 2016 06 08.
Article in English | MEDLINE | ID: mdl-27273450

ABSTRACT

Approximately 75% of melanomas have known driver oncogenic mutations in BRAF, NRAS, GNA11 or GNAQ, while the mutations providing constitutive oncogenic signaling in the remaining melanomas are not known. We established a melanoma cell line from a tumor with none of the common driver mutations. This cell line demonstrated a signaling profile similar to BRAF-mutants, but lacked sensitivity to the BRAF inhibitor vemurafenib. RNA-seq mutation data implicated CRAF R391W as the alternative driver mutation of this melanoma. CRAF R391W was homozygous and over expressed. These melanoma cells were highly sensitive to CRAF, but not BRAF knockdown. In reconstitution experiments, CRAF R391W, but not CRAF WT, transformed NIH3T3 cells in soft-agar colony formation assays, increased kinase activity in vitro, induced MAP kinase signaling and conferred vemurafenib resistance. MAP kinase inducing activity was dependent on CRAF dimerization. Thus, CRAF is a bona fide alternative oncogene for BRAF/NRAS/GNAQ/GNA11 wild type melanomas.


Subject(s)
Melanoma/genetics , Oncogenes , Proto-Oncogene Proteins c-raf/genetics , Aged , Animals , Humans , MAP Kinase Signaling System , Mice , NIH 3T3 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...