Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cancer Res ; 78(17): 5038-5049, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30026331

ABSTRACT

Regulatory T cells (Treg) are critical mediators of immunosuppression in established tumors, although little is known about their role in restraining immunosurveillance during tumorigenesis. Here, we employ an inducible autochthonous model of melanoma to investigate the earliest Treg and CD8 effector T-cell responses during oncogene-driven tumorigenesis. Induction of oncogenic BRAFV600E and loss of Pten in melanocytes led to localized accumulation of FoxP3+ Tregs, but not CD8 T cells, within 1 week of detectable increases in melanocyte differentiation antigen expression. Melanoma tumorigenesis elicited early expansion of shared tumor/self-antigen-specific, thymically derived Tregs in draining lymph nodes, and induced their subsequent recruitment to sites of tumorigenesis in the skin. Lymph node egress of tumor-activated Tregs was required for their C-C chemokine receptor 4 (Ccr4)-dependent homing to nascent tumor sites. Notably, BRAFV600E signaling controlled expression of Ccr4-cognate chemokines and governed recruitment of Tregs to tumor-induced skin sites. BRAFV600E expression alone in melanocytes resulted in nevus formation and associated Treg recruitment, indicating that BRAFV600E signaling is sufficient to recruit Tregs. Treg depletion liberated immunosurveillance, evidenced by CD8 T-cell responses against the tumor/self-antigen gp100, which was concurrent with the formation of microscopic neoplasia. These studies establish a novel role for BRAFV600E as a tumor cell-intrinsic mediator of immune evasion and underscore the critical early role of Treg-mediated suppression during autochthonous tumorigenesis.Significance: This work provides new insights into the mechanisms by which oncogenic pathways impact immune regulation in the nascent tumor microenvironment. Cancer Res; 78(17); 5038-49. ©2018 AACR.


Subject(s)
Carcinogenesis/genetics , Melanoma/genetics , Proto-Oncogene Proteins B-raf/genetics , T-Lymphocytes, Regulatory/metabolism , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Humans , Lymph Nodes/metabolism , Lymph Nodes/pathology , Melanocytes/immunology , Melanocytes/pathology , Melanoma/immunology , Melanoma/pathology , Mice , Mutation , PTEN Phosphohydrolase/genetics , Receptors, CCR4/genetics , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/genetics
2.
Sci Immunol ; 2(10)2017 Apr 14.
Article in English | MEDLINE | ID: mdl-28738020

ABSTRACT

Tissue-resident memory T (TRM) cells have been widely characterized in infectious disease settings; however, their role in mediating immunity to cancer remains unknown. We report that skin-resident memory T cell responses to melanoma are generated naturally as a result of autoimmune vitiligo. Melanoma antigen-specific TRM cells resided predominantly in melanocyte-depleted hair follicles and were maintained without recirculation or replenishment from the lymphoid compartment. These cells expressed CD103, CD69, and CLA (cutaneous lymphocyte antigen), but lacked PD-1 (programmed cell death protein-1) or LAG-3 (lymphocyte activation gene-3), and were capable of making IFN-γ (interferon-γ). CD103 expression on CD8 T cells was required for the establishment of TRM cells in the skin but was dispensable for vitiligo development. CD103+ CD8 TRM cells were critical for protection against melanoma rechallenge. This work establishes that CD103-dependent TRM cells play a key role in perpetuating antitumor immunity.

3.
Cancer Res ; 77(7): 1599-1610, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28202513

ABSTRACT

Acquired resistance to BRAFV600E inhibitors (BRAFi) in melanoma remains a common clinical obstacle, as is the case for any targeted drug therapy that can be developed given the plastic nature of cancers. Although there has been significant focus on the cancer cell-intrinsic properties of BRAFi resistance, the impact of BRAFi resistance on host immunity has not been explored. Here we provide preclinical evidence that resistance to BRAFi in an autochthonous mouse model of melanoma is associated with restoration of myeloid-derived suppressor cells (MDSC) in the tumor microenvironment, initially reduced by BRAFi treatment. In contrast to restoration of MDSCs, levels of T regulatory cells remained reduced in BRAFi-resistant tumors. Accordingly, tumor gene expression signatures specific for myeloid cell chemotaxis and homeostasis reappeared in BRAFi-resistant tumors. Notably, MDSC restoration relied upon MAPK pathway reactivation and downstream production of the myeloid attractant CCL2 in BRAFi-resistant melanoma cells. Strikingly, although combination checkpoint blockade (anti-CTLA-4 + anti-PD-1) was ineffective against BRAFi-resistant melanomas, the addition of MDSC depletion/blockade (anti-Gr-1 + CCR2 antagonist) prevented outgrowth of BRAFi-resistant tumors. Our results illustrate how extrinsic pathways of immunosuppression elaborated by melanoma cells dominate the tumor microenvironment and highlight the need to target extrinsic as well as intrinsic mechanisms of drug resistance. Cancer Res; 77(7); 1599-610. ©2017 AACR.


Subject(s)
Melanoma/drug therapy , Myeloid-Derived Suppressor Cells/physiology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Animals , Cell Line, Tumor , Chemokine CCL2/physiology , Drug Resistance, Neoplasm , Humans , Immunotherapy , Indoles/pharmacology , MAP Kinase Signaling System , Melanoma/immunology , Mice , Mice, Inbred C57BL , Receptors, CCR2/antagonists & inhibitors , Receptors, CCR2/physiology , Sulfonamides/pharmacology , T-Lymphocytes, Regulatory/physiology , Tumor Microenvironment
4.
Oncoimmunology ; 4(2): e988039, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25949884

ABSTRACT

As BRAFV600E-inhibitors become standard treatment for many metastatic melanoma patients, research has begun to elucidate their impact on the tumor immune landscape. Here, we highlight our recent studies demonstrating the ability of melanoma cell-intrinsic BRAFV600E-inhibition to selectively reduce intratumoral immunosuppressive cell populations and enhance antitumor CD8+ T-cell immunity.

5.
Cancer Immunol Res ; 2(11): 1044-50, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25183499

ABSTRACT

A growing body of evidence suggests that BRAF inhibitors, in addition to their acute tumor growth-inhibitory effects, can also promote immune responses to melanoma. The present study aimed to define the immunologic basis of BRAF-inhibitor therapy using the Braf/Pten model of inducible, autochthonous melanoma on a pure C57BL/6 background. In the tumor microenvironment, BRAF inhibitor PLX4720 functioned by on-target mechanisms to selectively decrease both the proportions and absolute numbers of CD4(+)Foxp3(+) regulatory T cells (Treg) and CD11b(+)Gr1(+) myeloid-derived suppressor cells (MDSC), while preserving numbers of CD8(+) effector T cells. In PLX4720-treated mice, the intratumoral Treg populations decreased significantly, demonstrating enhanced apopotosis. CD11b(+) myeloid cells from PLX4720-treated tumors also exhibited decreased immunosuppressive function on a per-cell basis. In accordance with a reversion of tumor immune suppression, tumors that had been treated with PLX4720 grew with reduced kinetics after treatment was discontinued, and this growth delay was dependent on CD8 T cells. These findings demonstrate that BRAF inhibition selectively reverses two major mechanisms of immunosuppression in melanoma and liberates host-adaptive antitumor immunity.


Subject(s)
Antineoplastic Agents/pharmacology , Indoles/pharmacology , Melanoma, Experimental/immunology , Myeloid Cells/drug effects , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Sulfonamides/pharmacology , T-Lymphocytes, Regulatory/drug effects , Animals , Cross-Priming , Enzyme Inhibitors/pharmacology , Flow Cytometry , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
7.
J Immunol ; 192(4): 1433-9, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24403535

ABSTRACT

Vitiligo is a CD8 T cell-mediated autoimmune disease that has been shown to promote the longevity of memory T cell responses to melanoma. However, mechanisms whereby melanocyte/melanoma Ag-specific T cell responses are perpetuated in the context of vitiligo are not well understood. These studies investigate the possible phenomenon of naive T cell priming in hosts with melanoma-initiated, self-perpetuating, autoimmune vitiligo. Using naive pmel (gp10025-33-specific) transgenic CD8 T cells, we demonstrate that autoimmune melanocyte destruction induces naive T cell proliferation in skin-draining lymph nodes, in an Ag-dependent fashion. These pmel T cells upregulate expression of CD44, P-selectin ligand, and granzyme B. However, they do not downregulate CD62L, nor do they acquire the ability to produce IFN-γ, indicating a lack of functional priming. Accordingly, adult thymectomized mice exhibit no reduction in the severity or kinetics of depigmentation or long-lived protection against melanoma, indicating that the continual priming of naive T cells is not required for vitiligo or its associated antitumor immunity. Despite this, depletion of CD4 T cells during the course of vitiligo rescues the priming of naive pmel T cells that are capable of producing IFN-γ and persisting as memory, suggesting an ongoing and dominant mechanism of suppression by regulatory T cells. This work reveals the complex regulation of self-reactive CD8 T cells in vitiligo and demonstrates the overall poorly immunogenic nature of this autoimmune disease setting.


Subject(s)
Autoimmune Diseases/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Melanoma/immunology , Vitiligo/immunology , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/transplantation , Cell Line, Tumor , Cell Proliferation , Disease Progression , Female , Granzymes/biosynthesis , Hyaluronan Receptors/biosynthesis , Hyaluronan Receptors/metabolism , Interferon-gamma/biosynthesis , Interferon-gamma/metabolism , L-Selectin/biosynthesis , L-Selectin/metabolism , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphocyte Activation/immunology , Male , Melanocytes/immunology , Mice , Mice, Inbred C57BL , P-Selectin/biosynthesis , P-Selectin/metabolism , Skin/immunology , Up-Regulation , gp100 Melanoma Antigen/genetics , gp100 Melanoma Antigen/metabolism
8.
J Neuroimmune Pharmacol ; 8(5): 1303-19, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24052414

ABSTRACT

JC virus (JCV) is a ubiquitous human polyomavirus that causes the demyelinating disease Progressive Multifocal Leukoencephalopathy (PML). JCV replicates in limited cell types in culture, predominantly in human glial cells. Following introduction of a replication defective SV40 mutant that expressed large T protein into a heterogeneous culture of human fetal brain cells, multiple phenotypes became immortalized (SVG cells). A subset of SVG cells could support JCV replication. In the current study, clonal cell lines were selected from the original SVG cell culture. The 5F4 clone showed low levels of viral growth. The 10B1 clone was highly permissive for JCV DNA replication and gene expression and supported persistent and stable JCV infection over months in culture. Microarray analysis revealed that viral infection did not significantly change gene expression in these cells. More resistant 5F4 cells expressed high levels of transcription factors known to inhibit JCV transcription. Interestingly, 5F4 cells expressed high levels of RNA of markers of radial glia and 10B1 cells had high expression of markers of immature glial cells and activation of transcription regulators important for stem/progenitor cell self-renewal. These SVG-derived clonal cell lines provide a biologically relevant model to investigate cell type differences in JCV host range and pathogenesis, as well as neural development. Several transcription regulators were identified which may be targets for therapeutic modulation of expression to abrogate JCV replication in PML patients. Additionally, these clonal cell lines can provide a consistent culture platform for testing therapies against JCV infection of the central nervous system.


Subject(s)
JC Virus/physiology , Neuroglia/virology , Polyomavirus Infections/genetics , Virus Replication/genetics , Cell Line , Clone Cells , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Humans , Immunoblotting , Intercellular Signaling Peptides and Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transcriptome
9.
PLoS One ; 6(10): e26491, 2011.
Article in English | MEDLINE | ID: mdl-22046294

ABSTRACT

BACKGROUND: We have previously demonstrated that temporary depletion of CD4 T cells in mice with progressive B16 melanoma, followed by surgical tumor excision, induces protective memory CD8 T cell responses to melanoma/melanocyte antigens. We also showed that persistence of these CD8 T cells is supported, in an antigen-dependent fashion, by concurrent autoimmune melanocyte destruction. Herein we explore the requirement of CD4 T cell help in priming and maintaining this protective CD8 T cell response to melanoma. METHODOLOGY AND PRINCIPAL FINDINGS: To induce melanoma/melanocyte antigen-specific CD8 T cells, B16 tumor bearing mice were depleted of regulatory T cells (T(reg)) by either temporary, or long-term continuous treatment with anti-CD4 (mAb clone GK1.5). Total depletion of CD4 T cells led to significant priming of IFN-γ-producing CD8 T cell responses to TRP-2 and gp100. Surprisingly, treatment with anti-CD25 (mAb clone PC61), to specifically deplete T(reg) cells while leaving help intact, was ineffective at priming CD8 T cells. Thirty to sixty days after primary tumors were surgically excised, mice completely lacking CD4 T cell help developed autoimmune vitiligo, and maintained antigen-specific memory CD8 T cell responses that were highly effective at producing cytokines (IFN-γ, TNF-α, and IL-2). Mice lacking total CD4 T cell help also mounted protection against re-challenge with B16 melanoma sixty days after primary tumor excision. CONCLUSIONS AND SIGNIFICANCE: This work establishes that CD4 T cell help is dispensable for the generation of protective memory T cell responses to melanoma. Our findings support further use of CD4 T cell depletion therapy for inducing long-lived immunity to cancer.


Subject(s)
Immunologic Memory/immunology , Melanoma, Experimental/immunology , T-Lymphocytes/immunology , Animals , Antigen Presentation , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/biosynthesis , Mice
10.
J Clin Invest ; 121(5): 1797-809, 2011 May.
Article in English | MEDLINE | ID: mdl-21540555

ABSTRACT

A link between autoimmunity and improved antitumor immunity has long been recognized, although the exact mechanistic relationship between these two phenomena remains unclear. In the present study we have found that vitiligo, the autoimmune destruction of melanocytes, generates self antigen required for mounting persistent and protective memory CD8+ T cell responses to melanoma. Vitiligo developed in approximately 60% of mice that were depleted of regulatory CD4+ T cells and then subjected to surgical excision of large established B16 melanomas. Mice with vitiligo generated 10-fold larger populations of CD8+ memory T cells specific for shared melanoma/melanocyte antigens. CD8+ T cells in mice with vitiligo acquired phenotypic and functional characteristics of effector memory, suggesting that they were supported by ongoing antigen stimulation. Such responses were not generated in melanocyte-deficient mice, indicating a requirement for melanocyte destruction in maintaining CD8+ T cell immunity to melanoma. Vitiligo-associated memory CD8+ T cells provided durable tumor protection, were capable of mounting a rapid recall response to melanoma, and did not demonstrate phenotypic or functional signs of exhaustion even after many months of exposure to antigen. This work establishes melanocyte destruction as a key determinant of lasting melanoma-reactive immune responses, thus illustrating that immune-mediated destruction of normal tissues can perpetuate adaptive immune responses to cancer.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Melanocytes/cytology , Melanoma/metabolism , Skin Neoplasms/metabolism , Animals , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Homeostasis , Immunologic Memory , Interferon-gamma/metabolism , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mice, Transgenic , Vitiligo/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL