Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Science ; 379(6627): 37-39, 2023 01 06.
Article in English | MEDLINE | ID: mdl-36603084

ABSTRACT

Highlights from the Science family of journals.

2.
Science ; 377(6609): 936-937, 2022 08 26.
Article in English | MEDLINE | ID: mdl-36007023

ABSTRACT

Highlights from the Science family of journals.

5.
Science ; 376(6592): 471-473, 2022 04 29.
Article in English | MEDLINE | ID: mdl-35482851

ABSTRACT

Highlights from the Science family of journals.

8.
Science ; 375(6587): 1368-1370, 2022 03 25.
Article in English | MEDLINE | ID: mdl-35324289

ABSTRACT

Highlights from the Science family of journals.

9.
Science ; 375(6586): 1241-1243, 2022 Mar 18.
Article in English | MEDLINE | ID: mdl-35298252

ABSTRACT

Highlights from the Science family of journals.

10.
Science ; 375(6582): 731-733, 2022 Feb 18.
Article in English | MEDLINE | ID: mdl-35175815

ABSTRACT

Highlights from the Science family of journals.

12.
13.
Science ; 374(6568): 704-706, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34735247

ABSTRACT

Highlights from the Science family of journals.

14.
Science ; 373(6559): 1100-1102, 2021 Sep 03.
Article in English | MEDLINE | ID: mdl-34516869

ABSTRACT

Highlights from the Science family of journals.

15.
J Virol ; 95(19): e0056621, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34260270

ABSTRACT

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.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Ectromelia, Infectious/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/immunology , Virus Diseases/immunology , Animals , Antigen-Presenting Cells/immunology , CD11 Antigens/analysis , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation , Core Binding Factor Alpha 3 Subunit/metabolism , Cytotoxicity, Immunologic , Ectromelia virus/physiology , Ectromelia, Infectious/virology , Histocompatibility Antigens Class II/analysis , Liver/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Spleen/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Th1 Cells/metabolism , Transcriptome , Virus Replication
16.
Cancer Immunol Res ; 8(9): 1114-1121, 2020 09.
Article in English | MEDLINE | ID: mdl-32661093

ABSTRACT

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.


Subject(s)
Acrylonitrile/analogs & derivatives , Aniline Compounds/therapeutic use , CD8-Positive T-Lymphocytes/drug effects , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Acrylonitrile/pharmacology , Acrylonitrile/therapeutic use , Aniline Compounds/pharmacology , Animals , Humans , Male , Mice
17.
Cell Rep ; 30(2): 510-524.e6, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31940493

ABSTRACT

Immune checkpoint inhibitors have improved patient survival in melanoma, but the innate resistance of many patients necessitates the investigation of alternative immune targets. Many immune checkpoint proteins lack proper characterization, including V-domain Ig suppressor of T cell activation (VISTA). VISTA expression on immune cells can suppress T cell activity; however, few studies have investigated its expression and regulation in cancer cells. In this study, we observe that VISTA is expressed in melanoma patient samples and cell lines. Tumor cell-specific expression of VISTA promotes tumor onset in vivo, associated with increased intratumoral T regulatory cells, and enhanced PDL-1 expression on tumor-infiltrating macrophages. VISTA transcript levels are regulated by the stemness factor Forkhead box D3 (FOXD3). BRAF inhibition upregulates FOXD3 and reduces VISTA expression. Overall, this study demonstrates melanoma cell expression of VISTA and its regulation by FOXD3, contributing to the rationale for therapeutic strategies that combine targeted inhibitors with immune checkpoint blockade.


Subject(s)
B7 Antigens/biosynthesis , Forkhead Transcription Factors/metabolism , Melanoma/genetics , Adult , Aged , Aged, 80 and over , Animals , B7 Antigens/genetics , B7 Antigens/immunology , B7 Antigens/metabolism , Cell Line, Tumor , Female , Forkhead Transcription Factors/genetics , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Male , Melanoma/immunology , Melanoma/metabolism , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Middle Aged , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Survival Analysis , T-Lymphocytes/immunology
18.
Br J Cancer ; 122(6): 789-800, 2020 03.
Article in English | MEDLINE | ID: mdl-31932756

ABSTRACT

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.


Subject(s)
Melanoma/drug therapy , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Animals , Humans , Mice , Mice, Nude , Protein Kinase Inhibitors/pharmacology , Transfection , Up-Regulation
19.
Cancer Discov ; 10(2): 254-269, 2020 02.
Article in English | MEDLINE | ID: mdl-31796433

ABSTRACT

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.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Melanoma/drug therapy , Proto-Oncogene Proteins B-raf/genetics , Pyroptosis/drug effects , Skin Neoplasms/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor/transplantation , Dendritic Cells/drug effects , Dendritic Cells/immunology , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/immunology , Female , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Male , Melanoma/genetics , Melanoma/immunology , Mice , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation , Proof of Concept Study , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/metabolism , Pyroptosis/genetics , Pyroptosis/immunology , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
20.
Pigment Cell Melanoma Res ; 33(2): 372-377, 2020 03.
Article in English | MEDLINE | ID: mdl-31696640

ABSTRACT

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.


Subject(s)
Aminopyridines/pharmacology , Macrophages/pathology , Melanoma/pathology , Proteins/antagonists & inhibitors , Pyrroles/pharmacology , Skin Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Macrophages/drug effects , Male , Mice, Inbred C57BL , Proteins/metabolism , Proto-Oncogene Proteins B-raf/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...