Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Cancer Discov ; 10(9): 1296-1311, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32371478

RESUMEN

The molecular mechanisms leading to resistance to PD-1 blockade are largely unknown. Here, we characterize tumor biopsies from a patient with melanoma who displayed heterogeneous responses to anti-PD-1 therapy. We observe that a resistant tumor exhibited a loss-of-function mutation in the tumor suppressor gene FBXW7, whereas a sensitive tumor from the same patient did not. Consistent with a functional role in immunotherapy response, inactivation of Fbxw7 in murine tumor cell lines caused resistance to anti-PD-1 in immunocompetent animals. Loss of Fbxw7 was associated with altered immune microenvironment, decreased tumor-intrinsic expression of the double-stranded RNA (dsRNA) sensors MDA5 and RIG1, and diminished induction of type I IFN and MHC-I expression. In contrast, restoration of dsRNA sensing in Fbxw7-deficient cells was sufficient to sensitize them to anti-PD-1. Our results thus establish a new role for the commonly inactivated tumor suppressor FBXW7 in viral sensing and sensitivity to immunotherapy. SIGNIFICANCE: Our findings establish a role of the commonly inactivated tumor suppressor FBXW7 as a genomic driver of response to anti-PD-1 therapy. Fbxw7 loss promotes resistance to anti-PD-1 through the downregulation of viral sensing pathways, suggesting that therapeutic reactivation of these pathways could improve clinical responses to checkpoint inhibitors in genomically defined cancer patient populations.This article is highlighted in the In This Issue feature, p. 1241.


Asunto(s)
Resistencia a Antineoplásicos/genética , Proteína 7 que Contiene Repeticiones F-Box-WD/genética , Inhibidores de Puntos de Control Inmunológico/farmacología , Neoplasias Cutáneas/tratamiento farmacológico , Anciano , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Línea Celular Tumoral/trasplante , Proteína 58 DEAD Box/genética , Proteína 58 DEAD Box/metabolismo , Modelos Animales de Enfermedad , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Regulación Neoplásica de la Expresión Génica/inmunología , Células HeLa , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Helicasa Inducida por Interferón IFIH1/genética , Helicasa Inducida por Interferón IFIH1/metabolismo , Mutación con Pérdida de Función , Masculino , Ratones , Mutagénesis Sitio-Dirigida , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , ARN Bicatenario/inmunología , ARN Bicatenario/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
2.
Nat Commun ; 10(1): 5157, 2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31727958

RESUMEN

Most targeted cancer therapies fail to achieve complete tumor regressions or attain durable remissions. To understand why these treatments fail to induce robust cytotoxic responses despite appropriately targeting oncogenic drivers, here we systematically interrogated the dependence of cancer cells on the BCL-2 family of apoptotic proteins after drug treatment. We observe that multiple targeted therapies, including BRAF or EGFR inhibitors, rapidly deplete the pro-apoptotic factor NOXA, thus creating a dependence on the anti-apoptotic protein MCL-1. This adaptation requires a pathway leading to destabilization of the NOXA mRNA transcript. We find that interruption of this mechanism of anti-apoptotic adaptive resistance dramatically increases cytotoxic responses in cell lines and a murine melanoma model. These results identify NOXA mRNA destabilization/MCL-1 adaptation as a non-genomic mechanism that limits apoptotic responses, suggesting that sequencing of MCL-1 inhibitors with targeted therapies could overcome such widespread and clinically important resistance.


Asunto(s)
Resistencia a Antineoplásicos/genética , Terapia Molecular Dirigida , Proteínas Proto-Oncogénicas c-bcl-2/genética , Estabilidad del ARN/genética , Animales , Apoptosis , Secuencia de Bases , Línea Celular Tumoral , Humanos , Masculino , Ratones Desnudos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Tristetraprolina/metabolismo
3.
Mol Cancer Ther ; 15(10): 2465-2474, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27507852

RESUMEN

Clear cell renal cell carcinoma (ccRCC) is characterized by intratumoral hypoxia and chemoresistance. The hypoxia-inducible factors HIF1α and HIF2α play a crucial role in ccRCC initiation and progression. We previously identified the sphingosine kinase 1/sphingosine 1-phosphate (SphK1/S1P) pathway as a new modulator of HIF1α and HIF2α under hypoxia in various cancer cell models. Here, we report that FTY720, an inhibitor of the S1P signaling pathway, inhibits both HIF1α and HIF2α accumulation in several human cancer cell lines. In a ccRCC heterotopic xenograft model, we show that FTY720 transiently decreases HIF1α and HIF2α intratumoral level and modifies tumor vessel architecture within 5 days of treatment, suggesting a vascular normalization. In mice bearing subcutaneous ccRCC tumor, FTY720 and a gemcitabine-based chemotherapy alone display a limited effect, whereas, in combination, there is a significant effect on tumor size without toxicity. Noteworthy, administration of FTY720 for 5 days before chemotherapy is not associated with a more effective tumor control, suggesting a mode of action mainly independent of the vascular remodeling. In conclusion, these findings demonstrate that FTY720 could successfully sensitize ccRCC to chemotherapy and establish this molecule as a potent therapeutic agent for ccRCC treatment, independently of drug scheduling. Mol Cancer Ther; 15(10); 2465-74. ©2016 AACR.


Asunto(s)
Antineoplásicos/farmacología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Clorhidrato de Fingolimod/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Transducción de Señal/efectos de los fármacos , Remodelación Vascular/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos , Femenino , Expresión Génica , Humanos , Hipoxia/genética , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Lisofosfolípidos , Ratones , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Consumo de Oxígeno , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Oncotarget ; 6(15): 13803-21, 2015 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-25915662

RESUMEN

Hypoxia promotes neovascularization, increased tumor growth, and therapeutic resistance. The transcription factor, hypoxia-inducible factor 1α (HIF-1α), has been reported as the master driver of adaptation to hypoxia. We previously identified the sphingosine kinase 1/sphingosine 1-phosphate (SphK1/S1P) pathway as a new modulator of HIF-1α under hypoxia. Taking advantage of a monoclonal antibody neutralizing extracellular S1P (sphingomab), we report that inhibition of S1P extracellular signaling blocks HIF-1α accumulation and activity in several cancer cell models exposed to hypoxia. In an orthotopic xenograft model of prostate cancer, we show that sphingomab reduces hypoxia and modifies vessel architecture within 5 days of treatment, leading to increased intratumoral blood perfusion. Supporting the notion that a transient vascular normalization of tumor vessels is the mechanism by which sphingomab exerts its effects, we demonstrate that administration of the antibody for 5 days before chemotherapy is more effective at local tumor control and metastatic dissemination than any other treatment scheduling. These findings validate sphingomab as a potential new normalization agent that could contribute to successful sensitization of hypoxic tumors to chemotherapy.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Lisofosfolípidos/antagonistas & inhibidores , Lisofosfolípidos/inmunología , Neoplasias de la Próstata/tratamiento farmacológico , Esfingosina/análogos & derivados , Animales , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Humanos , Masculino , Ratones , Ratones Desnudos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Neoplasias de la Próstata/irrigación sanguínea , Neoplasias de la Próstata/metabolismo , Distribución Aleatoria , Transducción de Señal , Esfingosina/antagonistas & inhibidores , Esfingosina/inmunología , Remodelación Vascular/efectos de los fármacos
5.
Mol Oncol ; 8(7): 1181-95, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24768038

RESUMEN

Sphingosine 1-phosphate (S1P) plays important roles in cell proliferation, differentiation or survival mainly through its surface G-protein-coupled receptors S1P1-5. Bone represents the major site of metastasis for prostate cancer (CaP) cells, which rely on bone-derived factors to support their proliferation and resistance to therapeutics. In the present work we have found that conditioned medium (CM) from the MC3T3 osteoblastic cell line or primary murine and human osteoblast-like cells, as well as co-culture with MC3T3 stimulate proliferation of CaP lines in S1P-dependent manner. In addition, osteoblastic-derived S1P induces resistance of CaP cells to therapeutics including chemotherapy and radiotherapy. When S1P release from osteoblastic cells is decreased (inhibition of SphK1, knock-down of SphK1 or the S1P transporter, Spns2 by siRNA) or secreted S1P neutralized with anti-S1P antibody, the proliferative and survival effects of osteoblasts on CaP cells are abolished. Because of the paracrine nature of the signaling, we studied the role of the S1P receptors expressed on CaP cells in the communication with S1P secreted by osteoblasts. Strategies aimed at down-regulating S1P1, S1P2 or S1P3 (siRNA, antagonists), established the exclusive role of the S1P/S1P1 signaling between osteoblasts and CaP cells. Bone metastases from CaP are associated with osteoblastic differentiation resulting in abnormal bone formation. We show that the autocrine S1P/S1P3 signaling is central during differentiation to mature osteoblasts by regulating Runx2 level, a key transcription factor involved in osteoblastic maturation. Importantly, differentiated osteoblasts exhibited enhanced secretion of S1P and further stimulated CaP cell proliferation in a S1P-dependent manner. By establishing the dual role of osteoblast-borne S1P on both osteoblastic differentiation and CaP cell proliferation and survival, we uncover the importance of S1P in the bone metastatic microenvironment, which may open a novel area of study for the treatment of CaP bone metastasis by targeting S1P.


Asunto(s)
Neoplasias Óseas/secundario , Lisofosfolípidos/metabolismo , Osteoblastos/patología , Próstata/patología , Neoplasias de la Próstata/patología , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Animales , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Neoplasias Óseas/terapia , Diferenciación Celular/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Técnicas de Cocultivo , Resistencia a Antineoplásicos , Humanos , Masculino , Ratones , Osteoblastos/citología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/terapia , Transducción de Señal , Esfingosina/metabolismo
6.
Adv Cancer Res ; 117: 117-41, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23290779

RESUMEN

Hypoxia, defined as a poor oxygenation, has been long recognized as a hallmark of solid tumors and a negative prognostic factor for response to therapeutics and survival of patients. Cancer cells have evolved biochemical mechanisms that allow them to react and adapt to hypoxia. At the cellular level, this adaptation is under the control of two related transcription factors, HIF-1 and HIF-2 (hypoxia-inducible factor), that respond rapidly to decreased oxygen levels to activate the expression of a broad range of genes promoting neoangiogenesis, glycolysis, metastasis, increased tumor growth, and resistance to treatments. Recent studies have identified the sphingosine kinase 1/sphingosine 1-phosphate (SphK1/S1P) signaling pathway-which elicits various cellular processes including cell proliferation, cell survival, or angiogenesis-as a new regulator of HIF-1 or HIF-2 activity. In this review, we will focus on how the inhibition/neutralization of the SphK1/S1P signaling could be exploited for cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Hipoxia , Lisofosfolípidos/metabolismo , Neoplasias/patología , Esfingosina/análogos & derivados , Animales , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neovascularización Patológica , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Transducción de Señal , Esfingosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA