Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
J Virol ; 95(19): e0056621, 2021 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-34260270

RESUMEN

Cytotoxic CD4 T lymphocytes (CD4-CTL) are important in antiviral immunity. For example, we have previously shown that in mice, CD4-CTL are important to control ectromelia virus (ECTV) infection. How viral infections induce CD4-CTL responses remains incompletely understood. We demonstrate here that not only ECTV but also vaccinia virus and lymphocytic choriomeningitis virus induce CD4-CTL, though the response to ECTV is stronger. Using ECTV, we also demonstrate that in contrast to CD8-CTL, CD4-CTL differentiation requires constant virus replication and ceases once the virus is controlled. We also show that major histocompatibility complex class II molecules on CD11c+ cells are required for CD4-CTL differentiation and for mousepox resistance. Transcriptional analysis indicated that antiviral CD4-CTL and noncytolytic T helper 1 (Th1) CD4 T cells have similar transcriptional profiles, suggesting that CD4-CTL are terminally differentiated classical Th1 cells. Interestingly, CD4-CTL and classical Th1 cells expressed similar mRNA levels of the transcription factors ThPOK and GATA-3, necessary for CD4 T cell linage commitment, and Runx3, required for CD8 T cell development and effector function. However, at the protein level, CD4-CTL had higher levels of the three transcription factors, suggesting that further posttranscriptional regulation is required for CD4-CTL differentiation. Finally, CRISPR/Cas9-mediated deletion of Runx3 in CD4 T cells inhibited CD4-CTL but not classical Th1 cell differentiation in response to ECTV infection. These results further our understanding of the mechanisms of CD4-CTL differentiation during viral infection and the role of posttranscriptionally regulated Runx3 in this process. IMPORTANCE While it is well established that cytotoxic CD4 T cells (CD4-CTLs) directly contribute to viral clearance, it remains unclear how CD4-CTL are induced. We now show that CD4-CTLs require sustained antigen presentation and are induced by CD11c-expressing antigen-presenting cells. Moreover, we show that CD4-CTLs are derived from the terminal differentiation of classical T helper 1 (Th1) subset of CD4 cells. Compared to Th1 cells, CD4-CTLs upregulate protein levels of the transcription factors ThPOK, Runx3, and GATA-3 posttranscriptionally. Deletion of Runx3 in differentiated CD4 T cells prevents induction of CD4-CTLs but not classical Th1 cells. These results advance our knowledge of how CD4-CTLs are induced during viral infection.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Ectromelia Infecciosa/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Citotóxicos/inmunología , Células TH1/inmunología , Virosis/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Antígenos CD11/análisis , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Citotoxicidad Inmunológica , Virus de la Ectromelia/fisiología , Ectromelia Infecciosa/virología , Antígenos de Histocompatibilidad Clase II/análisis , Hígado/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Bazo/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Citotóxicos/metabolismo , Células TH1/metabolismo , Transcriptoma , Replicación Viral
2.
Br J Cancer ; 122(6): 789-800, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31932756

RESUMEN

BACKGROUND: BRAF-mutant melanoma patients respond to BRAF inhibitors and MEK inhibitors (BRAFi/MEKi), but drug-tolerant cells persist, which may seed disease progression. Adaptive activation of receptor tyrosine kinases (RTKs) has been associated with melanoma cell drug tolerance following targeted therapy. While co-targeting individual RTKs can enhance the efficacy of BRAFi/MEKi effects, it remains unclear how to broadly target multiple RTKs to achieve more durable tumour growth inhibition. METHODS: The blockage of adaptive RTK responses by the new BET inhibitor (BETi), PLX51107, was measured by RPPA and Western blot. Melanoma growth was evaluated in vitro by colony assay and EdU staining, as well as in skin reconstructs, xenografts and PDX models following BRAFi, MEKi and/or PLX51107 treatment. RESULTS: Treatment with PLX51107 limited BRAFi/MEKi upregulation of ErbB3 and PDGFR-ß expression levels. Similar effects were observed following BRD2/4 depletion. In stage III melanoma patients, expression of BRD2/4 was strongly correlated with ErbB3. PLX51107 enhanced the effects of BRAFi/MEKi on inhibiting melanoma growth in vitro, in human skin reconstructs and in xenografts in vivo. Continuous triple drug combination treatment resulted in significant weight loss in mice, but intermittent BETi combined with continuous BRAFi/MEKi treatment was tolerable and improved durable tumour inhibition outcomes. CONCLUSIONS: Together, our data suggest that intermittent inhibition of BET proteins may improve the duration of responses following BRAFi/MEKi treatment in BRAF-mutant melanoma.


Asunto(s)
Melanoma/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Animales , Humanos , Ratones , Ratones Desnudos , Inhibidores de Proteínas Quinasas/farmacología , Transfección , Regulación hacia Arriba
3.
J Immunol ; 198(7): 2979-2988, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28202614

RESUMEN

It is well known that CD8+ tumor-infiltrating lymphocytes (TILs) are correlated with positive prognoses in cancer patients and are used to determine the efficacy of immune therapies. Although it is generally assumed that CD8+ TILs will be tumor-associated Ag (TAA) specific, it is unknown whether CD8+ T cells with specificity for common pathogens also infiltrate tumors. If so, the presence of these T cells could alter the interpretation of prognostic and diagnostic TIL assays. We compared TAA-specific and virus-specific CD8+ T cells in the same tumors using murine CMV, a herpesvirus that causes a persistent/latent infection, and vaccinia virus, a poxvirus that is cleared by the host. Virus-specific CD8+ TILs migrated into cutaneous melanoma lesions during acute infection with either virus, after a cleared vaccinia virus infection, and during a persistent/latent murine CMV infection. Virus-specific TILs developed independently of viral Ag in the tumor and, interestingly, expressed low or intermediate levels of full-length PD-1 in the tumor environment. Importantly, PD-1 expression could be markedly induced by Ag but did not correlate with dysfunction for virus-specific TILs, in sharp contrast to TAA-specific TILs in the same tumors. These data suggest that CD8+ TILs can reflect an individual's immune status, rather than exclusively representing TAA-specific T cells, and that PD-1 expression on CD8+ TILs is not always associated with repeated Ag encounter or dysfunction. Thus, functional virus-specific CD8+ TILs could skew the results of prognostic or diagnostic TIL assays.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma Experimental/inmunología , Virosis/complicaciones , Traslado Adoptivo , Animales , Antígenos de Neoplasias/inmunología , Citometría de Flujo , Infecciones por Herpesviridae/complicaciones , Infecciones por Herpesviridae/inmunología , Melanoma Experimental/complicaciones , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Muromegalovirus/inmunología , Reacción en Cadena de la Polimerasa , Receptor de Muerte Celular Programada 1/biosíntesis , Receptor de Muerte Celular Programada 1/inmunología , Vaccinia/complicaciones , Vaccinia/inmunología , Virus Vaccinia/inmunología , Virosis/inmunología
4.
Mol Ther ; 24(8): 1444-55, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27434584

RESUMEN

Cytomegalovirus is an attractive cancer vaccine platform because it induces strong, functional CD8(+) T-cell responses that accumulate over time and migrate into most tissues. To explore this, we used murine cytomegalovirus expressing a modified gp100 melanoma antigen. Therapeutic vaccination by the intraperitoneal and intradermal routes induced tumor infiltrating gp100-specific CD8(+) T-cells, but provided minimal benefit for subcutaneous lesions. In contrast, intratumoral infection of established tumor nodules greatly inhibited tumor growth and improved overall survival in a CD8(+) T-cell-dependent manner, even in mice previously infected with murine cytomegalovirus. Although murine cytomegalovirus could infect and kill B16F0s in vitro, infection was restricted to tumor-associated macrophages in vivo. Surprisingly, the presence of a tumor antigen in the virus only slightly increased the efficacy of intratumoral infection and tumor-specific CD8(+) T-cells in the tumor remained dysfunctional. Importantly, combining intratumoral murine cytomegalovirus infection with anti-PD-L1 therapy was synergistic, resulting in tumor clearance from over half of the mice and subsequent protection against tumor challenge. Thus, while a murine cytomegalovirus-based vaccine was poorly effective against established subcutaneous tumors, direct infection of tumor nodules unexpectedly delayed tumor growth and synergized with immune checkpoint blockade to promote tumor clearance and long-term protection.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Inmunidad , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Muromegalovirus/fisiología , Animales , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Terapia Combinada , Expresión Génica , Orden Génico , Genes Reporteros , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Inmunoterapia , Macrófagos/inmunología , Macrófagos/metabolismo , Melanoma Experimental/patología , Melanoma Experimental/terapia , Ratones , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Resultado del Tratamiento , Carga Tumoral , Vacunación , Antígeno gp100 del Melanoma/genética
5.
Science ; 379(6627): 37-39, 2023 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-36603084

RESUMEN

Highlights from the Science family of journals.

6.
Science ; 376(6592): 471-473, 2022 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-35482851

RESUMEN

Highlights from the Science family of journals.

7.
Science ; 375(6582): 731-733, 2022 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-35175815

RESUMEN

Highlights from the Science family of journals.

8.
Science ; 375(6587): 1368-1370, 2022 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-35324289

RESUMEN

Highlights from the Science family of journals.

9.
Science ; 377(6609): 936-937, 2022 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-36007023

RESUMEN

Highlights from the Science family of journals.

10.
11.
13.
14.
Science ; 375(6586): 1241-1243, 2022 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-35298252

RESUMEN

Highlights from the Science family of journals.

16.
Science ; 374(6568): 704-706, 2021 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-34735247

RESUMEN

Highlights from the Science family of journals.

17.
Science ; 373(6559): 1100-1102, 2021 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-34516869

RESUMEN

Highlights from the Science family of journals.

18.
Pigment Cell Melanoma Res ; 33(2): 372-377, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31696640

RESUMEN

Bromodomain and extra-terminal inhibitors (BETi) delay tumor growth, in part, through tumor cell intrinsic alterations and initiation of anti-tumor CD8+ T-cell responses. By contrast, BETi effects on pro-tumoral immune responses remain unclear. Here, we show that the next-generation BETi, PLX51107, delayed tumor growth to differing degrees in Braf V600E melanoma syngeneic mouse models. These differential responses were associated with the influx of tumor-associated macrophages during BETi treatment. Tumors that were poorly responsive to PLX51107 showed increased influx of colony-stimulating factor-1 receptor (CSF-1R)-positive tumor-associated macrophages. We depleted CSF-1R+ tumor-associated macrophages with the CSF-1R inhibitor, PLX3397, in combination with PLX51107. Treatment with PLX3397 enhanced the efficacy of PLX51107 in poorly responsive Braf V600E syngeneic melanomas in vivo. These findings suggest that tumor-associated macrophage accumulation limits BETi efficacy and that co-treatment with PLX3397 can improve response to PLX51107, offering a potential novel combination therapy for metastatic melanoma patients.


Asunto(s)
Aminopiridinas/farmacología , Macrófagos/patología , Melanoma/patología , Proteínas/antagonistas & inhibidores , Pirroles/farmacología , Neoplasias Cutáneas/patología , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Macrófagos/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Proteínas/metabolismo , Proteínas Proto-Oncogénicas B-raf/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Resultado del Tratamiento
19.
Cancer Discov ; 10(2): 254-269, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31796433

RESUMEN

Combinations of BRAF inhibitors and MEK inhibitors (BRAFi + MEKi) are FDA-approved to treat BRAF V600E/K-mutant melanoma. Efficacy of BRAFi + MEKi associates with cancer cell death and alterations in the tumor immune microenvironment; however, the links are poorly understood. We show that BRAFi + MEKi caused durable melanoma regression in an immune-mediated manner. BRAFi + MEKi treatment promoted cleavage of gasdermin E (GSDME) and release of HMGB1, markers of pyroptotic cell death. GSDME-deficient melanoma showed defective HMGB1 release, reduced tumor-associated T cell and activated dendritic cell infiltrates in response to BRAFi + MEKi, and more frequent tumor regrowth after drug removal. Importantly, BRAFi + MEKi-resistant disease lacked pyroptosis markers and showed decreased intratumoral T-cell infiltration but was sensitive to pyroptosis-inducing chemotherapy. These data implicate BRAFi + MEKi-induced pyroptosis in antitumor immune responses and highlight new therapeutic strategies for resistant melanoma. SIGNIFICANCE: Targeted inhibitors and immune checkpoint agents have advanced the care of patients with melanoma; however, detailed knowledge of the intersection between these two research areas is lacking. We describe a molecular mechanism of targeted inhibitor regulation of an immune-stimulatory form of cell death and provide a proof-of-principle salvage therapy concept for inhibitor-resistant melanoma.See related commentary by Smalley, p. 176.This article is highlighted in the In This Issue feature, p. 161.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Melanoma/tratamiento farmacológico , Proteínas Proto-Oncogénicas B-raf/genética , Piroptosis/efectos de los fármacos , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Línea Celular Tumoral/trasplante , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Resistencia a Antineoplásicos/inmunología , Femenino , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Masculino , Melanoma/genética , Melanoma/inmunología , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Mutación , Prueba de Estudio Conceptual , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/metabolismo , Piroptosis/genética , Piroptosis/inmunología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
20.
Cancer Immunol Res ; 8(9): 1114-1121, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32661093

RESUMEN

Concurrent MEK and CDK4/6 inhibition shows promise in clinical trials for patients with advanced-stage mutant BRAF/NRAS solid tumors. The effects of CDK4/6 inhibitor (CDK4/6i) in combination with BRAF/MEK-targeting agents on the tumor immune microenvironment are unclear, especially in melanoma, for which immune checkpoint inhibitors are effective in approximately 50% of patients. Here, we show that patients progressing on CDK4/6i/MEK pathway inhibitor combinations exhibit T-cell exclusion. We found that MEK and CDK4/6 targeting was more effective at delaying regrowth of mutant BRAF melanoma in immunocompetent versus immune-deficient mice. Although MEK inhibitor (MEKi) treatment increased tumor immunogenicity and intratumoral recruitment of CD8+ T cells, the main effect of CDK4/6i alone and in combination with MEKi was increased expression of CD137L, a T-cell costimulatory molecule on immune cells. Depletion of CD8+ T cells or blockade of the CD137 ligand-receptor interaction reduced time to regrowth of melanomas in the context of treatment with CDK4/6i plus MEKi treatment in vivo Together, our data outline an antitumor immune-based mechanism and show the efficacy of targeting both the MEK pathway and CDK4/6.


Asunto(s)
Acrilonitrilo/análogos & derivados , Compuestos de Anilina/uso terapéutico , Linfocitos T CD8-positivos/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Acrilonitrilo/farmacología , Acrilonitrilo/uso terapéutico , Compuestos de Anilina/farmacología , Animales , Humanos , Masculino , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA