Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Cell ; 175(4): 998-1013.e20, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30388456

RESUMEN

Treatment of cancer has been revolutionized by immune checkpoint blockade therapies. Despite the high rate of response in advanced melanoma, the majority of patients succumb to disease. To identify factors associated with success or failure of checkpoint therapy, we profiled transcriptomes of 16,291 individual immune cells from 48 tumor samples of melanoma patients treated with checkpoint inhibitors. Two distinct states of CD8+ T cells were defined by clustering and associated with patient tumor regression or progression. A single transcription factor, TCF7, was visualized within CD8+ T cells in fixed tumor samples and predicted positive clinical outcome in an independent cohort of checkpoint-treated patients. We delineated the epigenetic landscape and clonality of these T cell states and demonstrated enhanced antitumor immunity by targeting novel combinations of factors in exhausted cells. Our study of immune cell transcriptomes from tumors demonstrates a strategy for identifying predictors, mechanisms, and targets for enhancing checkpoint immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inmunoterapia/métodos , Melanoma/inmunología , Transcriptoma , Animales , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales Humanizados/farmacología , Antígenos CD/inmunología , Antineoplásicos Inmunológicos/inmunología , Antineoplásicos Inmunológicos/farmacología , Apirasa/antagonistas & inhibidores , Apirasa/inmunología , Línea Celular Tumoral , Humanos , Antígenos Comunes de Leucocito/antagonistas & inhibidores , Antígenos Comunes de Leucocito/inmunología , Melanoma/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Factor 1 de Transcripción de Linfocitos T/metabolismo
3.
Nature ; 487(7408): 500-4, 2012 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-22763439

RESUMEN

Drug resistance presents a challenge to the treatment of cancer patients. Many studies have focused on cell-autonomous mechanisms of drug resistance. By contrast, we proposed that the tumour micro-environment confers innate resistance to therapy. Here we developed a co-culture system to systematically assay the ability of 23 stromal cell types to influence the innate resistance of 45 cancer cell lines to 35 anticancer drugs. We found that stroma-mediated resistance is common, particularly to targeted agents. We characterized further the stroma-mediated resistance of BRAF-mutant melanoma to RAF inhibitors because most patients with this type of cancer show some degree of innate resistance. Proteomic analysis showed that stromal cell secretion of hepatocyte growth factor (HGF) resulted in activation of the HGF receptor MET, reactivation of the mitogen-activated protein kinase (MAPK) and phosphatidylinositol-3-OH kinase (PI(3)K)-AKT signalling pathways, and immediate resistance to RAF inhibition. Immunohistochemistry experiments confirmed stromal cell expression of HGF in patients with BRAF-mutant melanoma and showed a significant correlation between HGF expression by stromal cells and innate resistance to RAF inhibitor treatment. Dual inhibition of RAF and either HGF or MET resulted in reversal of drug resistance, suggesting RAF plus HGF or MET inhibitory combination therapy as a potential therapeutic strategy for BRAF-mutant melanoma. A similar resistance mechanism was uncovered in a subset of BRAF-mutant colorectal and glioblastoma cell lines. More generally, this study indicates that the systematic dissection of interactions between tumours and their micro-environment can uncover important mechanisms underlying drug resistance.


Asunto(s)
Resistencia a Antineoplásicos , Factor de Crecimiento de Hepatocito/metabolismo , Melanoma/metabolismo , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Microambiente Tumoral/fisiología , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica , Línea Celular Tumoral , Técnicas de Cocultivo , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Indoles/farmacología , Indoles/uso terapéutico , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/patología , Terapia Molecular Dirigida , Mutación , Fosfatidilinositol 3-Quinasas/metabolismo , Pronóstico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteómica , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/metabolismo , Transducción de Señal/efectos de los fármacos , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , Vemurafenib
4.
Nucleic Acids Res ; 39(4): 1326-35, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21030442

RESUMEN

Promoter hypermethylation and heterochromatinization is a frequent event leading to gene inactivation and tumorigenesis. At the molecular level, inactivation of tumor suppressor genes in cancer has many similarities to the inactive X chromosome in female cells and is defined and maintained by DNA methylation and characteristic histone modifications. In addition, the inactive-X is marked by the histone macroH2A, a variant of H2A with a large non-histone region of unknown function. Studying tumor suppressor genes (TSGs) silenced in cancer cell lines, we find that when active, these promoters are associated with H2A.Z but become enriched for macroH2A1 once silenced. Knockdown of macroH2A1 was not sufficient for reactivation of silenced genes. However, when combined with DNA demethylation, macroH2A1 deficiency significantly enhanced reactivation of the tumor suppressor genes p16, MLH1 and Timp3 and inhibited cell proliferation. Our findings link macroH2A1 to heterochromatin of epigenetically silenced cancer genes and indicate synergism between macroH2A1 and DNA methylation in maintenance of the silenced state.


Asunto(s)
Metilación de ADN , Silenciador del Gen , Genes p16 , Histonas/metabolismo , Alelos , Línea Celular , Línea Celular Tumoral , Neoplasias del Colon/genética , Técnicas de Silenciamiento del Gen , Genes Supresores de Tumor , Histonas/genética , Humanos , Regiones Promotoras Genéticas
5.
Cell Rep Med ; 3(2): 100500, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35243413

RESUMEN

Immune checkpoint blockade (CPB) improves melanoma outcomes, but many patients still do not respond. Tumor mutational burden (TMB) and tumor-infiltrating T cells are associated with response, and integrative models improve survival prediction. However, integrating immune/tumor-intrinsic features using data from a single assay (DNA/RNA) remains underexplored. Here, we analyze whole-exome and bulk RNA sequencing of tumors from new and published cohorts of 189 and 178 patients with melanoma receiving CPB, respectively. Using DNA, we calculate T cell and B cell burdens (TCB/BCB) from rearranged TCR/Ig sequences and find that patients with TMBhigh and TCBhigh or BCBhigh have improved outcomes compared to other patients. By combining pairs of immune- and tumor-expressed genes, we identify three gene pairs associated with response and survival, which validate in independent cohorts. The top model includes lymphocyte-expressed MAP4K1 and tumor-expressed TBX3. Overall, RNA or DNA-based models combining immune and tumor measures improve predictions of melanoma CPB outcomes.


Asunto(s)
Melanoma , Transcriptoma , Humanos , Melanoma/tratamiento farmacológico , ARN , Análisis de Secuencia de ARN , Transcriptoma/genética , Secuenciación del Exoma
6.
Clin Cancer Res ; 24(19): 4771-4784, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29563139

RESUMEN

Purpose: Telomerase promoter mutations are highly prevalent in human tumors including melanoma. A subset of patients with metastatic melanoma often fail multiple therapies, and there is an unmet and urgent need to prolong disease control for those patients.Experimental Design: Numerous preclinical therapy-resistant models of human and mouse melanoma were used to test the efficacy of a telomerase-directed nucleoside, 6-thio-2'-deoxyguanosine (6-thio-dG). Integrated transcriptomics and proteomics approaches were used to identify genes and proteins that were significantly downregulated by 6-thio-dG.Results: We demonstrated the superior efficacy of 6-thio-dG both in vitro and in vivo that results in telomere dysfunction, leading to apoptosis and cell death in various preclinical models of therapy-resistant melanoma cells. 6-thio-dG concomitantly induces telomere dysfunction and inhibits the expression level of AXL.Conclusions: In summary, this study shows that indirectly targeting aberrant telomerase in melanoma cells with 6-thio-dG is a viable therapeutic approach in prolonging disease control and overcoming therapy resistance. Clin Cancer Res; 24(19); 4771-84. ©2018 AACR See related commentary by Teh and Aplin, p. 4629.


Asunto(s)
Desoxiguanosina/análogos & derivados , Melanoma/tratamiento farmacológico , Regiones Promotoras Genéticas/genética , Telomerasa/genética , Tionucleósidos/farmacología , Animales , Línea Celular Tumoral , Desoxiguanosina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Melanoma/genética , Melanoma/patología , Ratones , Mutación , Telómero/efectos de los fármacos , Telómero/genética
7.
Cancer Discov ; 8(2): 196-215, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29101162

RESUMEN

Ex vivo systems that incorporate features of the tumor microenvironment and model the dynamic response to immune checkpoint blockade (ICB) may facilitate efforts in precision immuno-oncology and the development of effective combination therapies. Here, we demonstrate the ability to interrogate ex vivo response to ICB using murine- and patient-derived organotypic tumor spheroids (MDOTS/PDOTS). MDOTS/PDOTS isolated from mouse and human tumors retain autologous lymphoid and myeloid cell populations and respond to ICB in short-term three-dimensional microfluidic culture. Response and resistance to ICB was recapitulated using MDOTS derived from established immunocompetent mouse tumor models. MDOTS profiling demonstrated that TBK1/IKKε inhibition enhanced response to PD-1 blockade, which effectively predicted tumor response in vivo Systematic profiling of secreted cytokines in PDOTS captured key features associated with response and resistance to PD-1 blockade. Thus, MDOTS/PDOTS profiling represents a novel platform to evaluate ICB using established murine models as well as clinically relevant patient specimens.Significance: Resistance to PD-1 blockade remains a challenge for many patients, and biomarkers to guide treatment are lacking. Here, we demonstrate feasibility of ex vivo profiling of PD-1 blockade to interrogate the tumor immune microenvironment, develop therapeutic combinations, and facilitate precision immuno-oncology efforts. Cancer Discov; 8(2); 196-215. ©2017 AACR.See related commentary by Balko and Sosman, p. 143See related article by Deng et al., p. 216This article is highlighted in the In This Issue feature, p. 127.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Animales , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Citocinas/metabolismo , Resistencia a Antineoplásicos , Citometría de Flujo , Humanos , Inmunohistoquímica , Inmunofenotipificación , Ratones , Técnicas Analíticas Microfluídicas , Receptor de Muerte Celular Programada 1/metabolismo , Esferoides Celulares , Imagen de Lapso de Tiempo , Células Tumorales Cultivadas
8.
Science ; 357(6356): 1156-1160, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28912244

RESUMEN

Growing evidence suggests that microbes can influence the efficacy of cancer therapies. By studying colon cancer models, we found that bacteria can metabolize the chemotherapeutic drug gemcitabine (2',2'-difluorodeoxycytidine) into its inactive form, 2',2'-difluorodeoxyuridine. Metabolism was dependent on the expression of a long isoform of the bacterial enzyme cytidine deaminase (CDDL), seen primarily in Gammaproteobacteria. In a colon cancer mouse model, gemcitabine resistance was induced by intratumor Gammaproteobacteria, dependent on bacterial CDDL expression, and abrogated by cotreatment with the antibiotic ciprofloxacin. Gemcitabine is commonly used to treat pancreatic ductal adenocarcinoma (PDAC), and we hypothesized that intratumor bacteria might contribute to drug resistance of these tumors. Consistent with this possibility, we found that of the 113 human PDACs that were tested, 86 (76%) were positive for bacteria, mainly Gammaproteobacteria.


Asunto(s)
Antimetabolitos Antineoplásicos/uso terapéutico , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/microbiología , Desoxicitidina/análogos & derivados , Resistencia a Antineoplásicos , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/microbiología , Animales , Neoplasias del Colon/microbiología , Desoxicitidina/uso terapéutico , Gammaproteobacteria/aislamiento & purificación , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Mycoplasma hyorhinis/aislamiento & purificación , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/microbiología , Gemcitabina , Neoplasias Pancreáticas
9.
Nat Commun ; 8(1): 1136, 2017 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-29070816

RESUMEN

Treatment with immune checkpoint blockade (CPB) therapies often leads to prolonged responses in patients with metastatic melanoma, but the common mechanisms of primary and acquired resistance to these agents remain incompletely characterized and have yet to be validated in large cohorts. By analyzing longitudinal tumor biopsies from 17 metastatic melanoma patients treated with CPB therapies, we observed point mutations, deletions or loss of heterozygosity (LOH) in beta-2-microglobulin (B2M), an essential component of MHC class I antigen presentation, in 29.4% of patients with progressing disease. In two independent cohorts of melanoma patients treated with anti-CTLA4 and anti-PD1, respectively, we find that B2M LOH is enriched threefold in non-responders (~30%) compared to responders (~10%) and associated with poorer overall survival. Loss of both copies of B2M is found only in non-responders. B2M loss is likely a common mechanism of resistance to therapies targeting CTLA4 or PD1.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Presentación de Antígeno/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Melanoma/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/inmunología , Presentación de Antígeno/genética , Antígeno CTLA-4/inmunología , Resistencia a Antineoplásicos/genética , Femenino , Humanos , Pérdida de Heterocigocidad , Melanoma/genética , Melanoma/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Metástasis de la Neoplasia , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Mutación Puntual , Receptor de Muerte Celular Programada 1/inmunología , Microglobulina beta-2/genética
10.
Cell Rep ; 21(7): 1953-1967, 2017 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-29141225

RESUMEN

Therapy of advanced melanoma is changing dramatically. Following mutational and biological subclassification of this heterogeneous cancer, several targeted and immune therapies were approved and increased survival significantly. To facilitate further advancements through pre-clinical in vivo modeling, we have established 459 patient-derived xenografts (PDX) and live tissue samples from 384 patients representing the full spectrum of clinical, therapeutic, mutational, and biological heterogeneity of melanoma. PDX have been characterized using targeted sequencing and protein arrays and are clinically annotated. This exhaustive live tissue resource includes PDX from 57 samples resistant to targeted therapy, 61 samples from responders and non-responders to immune checkpoint blockade, and 31 samples from brain metastasis. Uveal, mucosal, and acral subtypes are represented as well. We show examples of pre-clinical trials that highlight how the PDX collection can be used to develop and optimize precision therapies, biomarkers of response, and the targeting of rare genetic subgroups.


Asunto(s)
Xenoinjertos/patología , Melanoma/patología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Animales , Células Cultivadas , Xenoinjertos/metabolismo , Humanos , Melanoma/clasificación , Melanoma/genética , Ratones
11.
Cancer Discov ; 4(7): 816-27, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24771846

RESUMEN

UNLABELLED: Most melanomas harbor oncogenic BRAF(V600) mutations, which constitutively activate the MAPK pathway. Although MAPK pathway inhibitors show clinical benefit in BRAF(V600)-mutant melanoma, it remains incompletely understood why 10% to 20% of patients fail to respond. Here, we show that RAF inhibitor-sensitive and inhibitor-resistant BRAF(V600)-mutant melanomas display distinct transcriptional profiles. Whereas most drug-sensitive cell lines and patient biopsies showed high expression and activity of the melanocytic lineage transcription factor MITF, intrinsically resistant cell lines and biopsies displayed low MITF expression but higher levels of NF-κB signaling and the receptor tyrosine kinase AXL. In vitro, these MITF-low/NF-κB-high melanomas were resistant to inhibition of RAF and MEK, singly or in combination, and ERK. Moreover, in cell lines, NF-κB activation antagonized MITF expression and induced both resistance marker genes and drug resistance. Thus, distinct cell states characterized by MITF or NF-κB activity may influence intrinsic resistance to MAPK pathway inhibitors in BRAF(V600)-mutant melanoma. SIGNIFICANCE: Although most BRAF(V600)-mutant melanomas are sensitive to RAF and/or MEK inhibitors, a subset fails to respond to such treatment. This study characterizes a transcriptional cell state distinction linked to MITF and NF-κB that may modulate intrinsic sensitivity of melanomas to MAPK pathway inhibitors.


Asunto(s)
Resistencia a Antineoplásicos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Melanoma/genética , Melanoma/patología , Subunidad p50 de NF-kappa B/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/genética , Anilidas/farmacología , Bencimidazoles/farmacología , Benzocicloheptenos/farmacología , Línea Celular Tumoral , Células Cultivadas , Resistencia a Antineoplásicos/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Indoles/farmacología , Melanocitos/citología , Melanocitos/efectos de los fármacos , Melanocitos/metabolismo , Melanoma/tratamiento farmacológico , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción Asociado a Microftalmía/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Piridinas/farmacología , Quinolinas/farmacología , Sulfonamidas/farmacología , Triazoles/farmacología
12.
PLoS One ; 6(2): e17262, 2011 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-21364955

RESUMEN

MacroH2A1 is a histone H2A variant which contains a large non-histone C-terminal region of largely unknown function. Within this region is a macro domain which can bind ADP-ribose and related molecules. Most studies of macroH2A1 focus on the involvement of this variant in transcriptional repression. Studies in mouse embryos and in embryonic stem cells suggested that during early development macroH2A can be found at the centrosome. Centrosomal localization of macroH2A was later reported in somatic cells. Here we provide data showing that macroH2A1 does not localize to the centrosome and that the centrosomal signal observed with antibodies directed against the macroH2A1 non-histone region may be the result of antibody cross-reactivity.


Asunto(s)
Centrosoma/metabolismo , Histonas/genética , Histonas/metabolismo , Animales , Células Cultivadas , Cromatina/metabolismo , Técnicas de Silenciamiento del Gen , Variación Genética/fisiología , Proteínas Fluorescentes Verdes/metabolismo , Histonas/antagonistas & inhibidores , Humanos , Ratones , Proteínas Mutantes/metabolismo , Transporte de Proteínas/genética , Transporte de Proteínas/fisiología , Proteínas Recombinantes de Fusión/metabolismo , Distribución Tisular , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA