Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
1.
Clin Cancer Res ; 28(12): 2555-2566, 2022 06 13.
Article in English | MEDLINE | ID: mdl-35421231

ABSTRACT

PURPOSE: Immunotherapy for hepatocellular carcinoma (HCC) shows considerable promise in improving clinical outcomes. HepaVac-101 represents a single-arm, first-in-human phase I/II multicenter cancer vaccine trial for HCC (NCT03203005). It combines multipeptide antigens (IMA970A) with the TLR7/8/RIG I agonist CV8102. IMA970A includes 5 HLA-A*24 and 7 HLA-A*02 as well as 4 HLA-DR restricted peptides selected after mass spectrometric identification in human HCC tissues or cell lines. CV8102 is an RNA-based immunostimulator inducing a balanced Th1/Th2 immune response. PATIENTS AND METHODS: A total of 82 patients with very early- to intermediate-stage HCCs were enrolled and screened for suitable HLA haplotypes and 22 put on study treatment. This consisted in a single infusion of low-dose cyclophosphamide followed by nine intradermal coadministrations of IMA970A and CV8102. Only patients with no disease relapse after standard-of-care treatments were vaccinated. The primary endpoints of the HepaVac-101 clinical trial were safety, tolerability, and antigen-specific T-cell responses. Secondary or exploratory endpoints included additional immunologic parameters and survival endpoints. RESULTS: The vaccination showed a good safety profile. Transient mild-to-moderate injection-site reactions were the most frequent IMA970A/CV8102-related side effects. Immune responses against ≥1 vaccinated HLA class I tumor-associated peptide (TAA) and ≥1 vaccinated HLA class II TAA were respectively induced in 37% and 53% of the vaccinees. CONCLUSIONS: Immunotherapy may provide a great improvement in treatment options for HCC. HepaVac-101 is a first-in-human clinical vaccine trial with multiple novel HLA class I- and class II-restricted TAAs against HCC. The results are initial evidence for the safety and immunogenicity of the vaccine. Further clinical evaluations are warranted.


Subject(s)
Cancer Vaccines , Carcinoma, Hepatocellular , Liver Neoplasms , Adjuvants, Immunologic , Cancer Vaccines/adverse effects , Carcinoma, Hepatocellular/drug therapy , HLA-A Antigens , Humans , Immunotherapy/methods , Liver Neoplasms/drug therapy , Peptides
2.
PLoS One ; 11(7): e0160538, 2016.
Article in English | MEDLINE | ID: mdl-27467222

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0150623.].

3.
Oncoimmunology ; 5(4): e947175, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27141371

ABSTRACT

A strategic challenge facing clinicians treating patients afflicted with non-small cell lung cancer (NSCLC) is the development of approaches that combine conventional and novel therapies, including targeted therapies and immunotherapeutics. In a recent study, we explored the correlation between the expression of the tumor antigen family MAGE-A and the presence of EGFR and KRAS gene mutations in a large cohort of resected NSCLC patient specimens.

4.
PLoS One ; 11(3): e0150623, 2016.
Article in English | MEDLINE | ID: mdl-26937656

ABSTRACT

Immunotherapy approaches using checkpoint blockade, alone, or in combination with tumor antigen vaccination, or adoptive cell transfer, are emerging as promising approaches for the treatment of non-small cell lung cancer (NSCLC). In preparation for upcoming combined immunotherapy approaches in NSCLC, here, we have assessed spontaneous immune responses to XAGE-1b, a tumor specific antigen of the Cancer Testis Antigen group that has been previously reported to be spontaneously immunogenic in the Japanese population, in a cohort of Caucasian patients with NSCLC. We found spontaneous serological responses to XAGE-1b in 9% of the patients. Importantly, these responses were limited to, and represented 13% of, patients with adenocarcinoma tumors, the most frequent histological subtype, for which immunotherapy approaches are under development. Using a set of overlapping peptides spanning the entire XAGE-1b protein, and in support of the serological data, we detected significant XAGE-1b specific CD4+ T cell responses in all XAGE-1b seropositive patients and identified several CD4+ T cell epitopes. Altogether, our results support the relevance of the XAGE-1b antigen in Caucasians NSCLC patients with adenocarcinoma, and the implementation of future immunotherapies exploiting the high immunogenicity of the antigen in this patient population.


Subject(s)
Adenocarcinoma/therapy , Antigens, Neoplasm/immunology , Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/therapy , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Amino Acid Sequence , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/genetics , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cancer Vaccines/administration & dosage , Cancer Vaccines/biosynthesis , Cancer Vaccines/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Case-Control Studies , Epitopes/chemistry , Epitopes/immunology , Gene Expression , Humans , Immunotherapy, Active/methods , Interferon-gamma/agonists , Interferon-gamma/biosynthesis , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Molecular Sequence Data , Peptides/administration & dosage , Peptides/chemistry , Peptides/immunology , Primary Cell Culture
5.
PLoS One ; 10(12): e0145788, 2015.
Article in English | MEDLINE | ID: mdl-26709703

ABSTRACT

CD4 T cells secreting IL-10 or IL-17 are frequent at mucosal sites, where their equilibrium is important for simultaneously maintaining tolerance and immunity to the resident microbiota. The mode of action of these cells, however, is as yet incompletely understood. In this study, we have combined ex vivo analysis of CD4 T cells producing IL-10 or/and IL-17 with assessment of clonal populations isolated ex vivo using a cytokine catch assay. We found that circulating CD4 T cells secreting IL-10 or/and IL-17 ex vivo include both conventional FOXP3- CD4 T cells and FOXP3+ Helios- Treg. Upon assessment of clonal populations derived from single ex vivo isolated cytokine secreting cells, we found that IL-10 or/and IL-17 secreting cells prevalently secrete one or the other cytokine depending on the type of stimulation, the time after stimulation and the presence of microbial products. Namely, IL-10 secretion by clonal cells was prevalent at early time points after TCR mediated stimulation, was independent of co-stimulation and was increased in the presence of the microbial fermentation product butyrate. In contrast, IL-17 secretion was higher at later time points after TCR mediated stimulation and in the presence of co-stimulatory signals. Taken together, these results provide insights into the mechanisms that, through modulation of cytokine secretion depending on conditions, allow IL-10 and IL-17 producing CD4 T cells to contribute to maintain tolerance to microbes locally, while retaining the ability to participate in protective immune responses at distant sites.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Interleukin-10/metabolism , Interleukin-17/metabolism , Butyric Acid/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Forkhead Transcription Factors/metabolism , Gastrointestinal Microbiome/immunology , Humans , Ikaros Transcription Factor/metabolism , Immune Tolerance , Immunologic Memory , In Vitro Techniques , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
6.
Oncoimmunology ; 4(12): e946370, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26587312

ABSTRACT

In a recent study, we have demonstrated that T helper type 1 (TH1) cells specific for the tumor antigen NY-ESO-1 are amplified at ovarian tumor sites but are not "perverted" into immunosuppressive FOXP3+ regulatory T cells (Tregs). These findings encourage the development of protocols aiming to eliminate, or inactivate, FOXP3+ Tregs and reinforce Type I anticancer immunity, to improve clinical outcomes.

7.
Oncoimmunology ; 4(4): e998538, 2015 Apr.
Article in English | MEDLINE | ID: mdl-26137416

ABSTRACT

Whereas preclinical investigations and clinical studies have established that CD8+ T cells can profoundly affect cancer progression, the underlying mechanisms are still elusive. Challenging the prevalent view that the beneficial effect of CD8+ T cells in cancer is solely attributable to their cytotoxic activity, several reports have indicated that the ability of CD8+ T cells to promote tumor regression is dependent on their cytokine secretion profile and their ability to self-renew. Evidence has also shown that the tumor microenvironment can disarm CD8+ T cell immunity, leading to the emergence of dysfunctional CD8+ T cells. The existence of different types of CD8+ T cells in cancer calls for a more precise definition of the CD8+ T cell immune phenotypes in cancer and the abandonment of the generic terms "pro-tumor" and "antitumor." Based on recent studies investigating the functions of CD8+ T cells in cancer, we here propose some guidelines to precisely define the functional states of CD8+ T cells in cancer.

8.
Eur J Immunol ; 45(2): 474-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25446972

ABSTRACT

IL4I1 encodes an L-phenylalanine oxidase that inhibits T-cell proliferation. It has been recently reported that IL4I1 is expressed in TH17 cells as part of a mechanism that limits their pathogenicity. We have previously identified a population of human FOXP3(+) Treg cells that secrete IL-17 ex vivo; here, we addressed the expression of IL4I1 in that Treg-cell population. We found that in ex vivo isolated circulating Treg cells, IL4I1 expression is induced by activation. Moreover, IL4I1 expression is restricted to cells that do not express Helios, a transcription factor that characterizes natural Treg cells, but that express Aiolos, which is involved in the differentiation of TH17 and induced Treg cells. We also showed that conversion of Treg cells under inflammatory conditions increases IL4I1 expression, likely as part of a regulatory loop that attempts to limit the pathogenicity resulting from their conversion into TH17. The specific expression of IL4I1 in TH17 and iTreg cells may provide insights into approaches that aim at modulating these populations in different pathological conditions involving inflammation-mediated immunosuppression.


Subject(s)
Forkhead Transcription Factors/genetics , Ikaros Transcription Factor/genetics , L-Amino Acid Oxidase/genetics , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism , Cell Communication , Cell Differentiation , Forkhead Transcription Factors/immunology , Gene Expression Regulation/immunology , Humans , Ikaros Transcription Factor/immunology , Immune Tolerance , Interleukin-17/genetics , Interleukin-17/immunology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Ionomycin/pharmacology , L-Amino Acid Oxidase/immunology , Lymphocyte Activation , Signal Transduction , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Tetradecanoylphorbol Acetate/pharmacology , Th17 Cells/cytology , Th17 Cells/drug effects , Th17 Cells/immunology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/immunology
10.
Cancer Immunol Res ; 2(10): 943-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24866168

ABSTRACT

Non-small cell lung cancer (NSCLC) is a major public health problem, accounting for more cancer-related deaths than any other cancer. Both immunotherapy, based on the expression of tumor-specific antigens, and targeted therapy, based on the presence of oncogenic mutations, are under development for NSCLC. In this study, we analyzed the expression of MAGE-A, a cancer-testis antigen, in tumors from a cohort of patients with resected NSCLC with respect to their clinicopathologic characteristics and their mutational status for the EGFR and KRAS genes. We found MAGE-A expression by IHC in 43% of the tumors. MAGE-A expression was significantly more frequent in squamous tumors than in adenocarcinomas, did not correlate with disease stage, but was correlated significantly with high tumor grade and worse survival. EGFR and KRAS mutations were present in adenocarcinomas, but not in squamous tumors. Whereas the presence of EGFR mutations did not seem to affect survival, the presence of KRAS mutations was associated with early-stage disease and better survival. MAGE-A expression was absent from adenocarcinomas with KRAS mutations, but not significantly different in tumors with or without EGFR mutations. Together, the reported results provide guidance for the design of combination therapies in patients with NSCLC.


Subject(s)
Antigens, Neoplasm/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , ErbB Receptors/genetics , Lung Neoplasms/metabolism , Proto-Oncogene Proteins/genetics , ras Proteins/genetics , Aged , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/surgery , DNA Mutational Analysis/methods , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/surgery , Male , Middle Aged , Mutation , Neoplasm Grading , Neoplasm Staging , Proto-Oncogene Proteins p21(ras) , Survival Analysis
11.
J Immunother ; 37(2): 73-6, 2014.
Article in English | MEDLINE | ID: mdl-24509169

ABSTRACT

The cancer testis antigen (CTA), melanoma-associated antigen A3/6 (MAGE-A3/6), is expressed in human cancers of different histologic types, to variable extents, and is an important target for immunotherapy. In this study, to address the potential of MAGE-A3/6 as an immunotherapeutic target in breast cancer (BC), we assessed MAGE-A3/6 expression by PCR in a cohort of 362 primary BC tumors and analyzed the correlation between MAGE-A3/6 expression, tumors hormone receptor (HR) status, and other clinicopathologic features. We found expression of MAGE-A3/6 in 10% of primary BC tumors. MAGE-A3/6 expression was significantly correlated with estrogen receptor (ER) and progesterone receptor (PR) negative status and was frequent in ER (29%) and in PR (24%) tumors. MAGE-A3/6 expression was also significantly associated with high histologic grade but not with patients age, tumor size, tumor type, lymph-node invasion, and human epidermal growth factor receptor 2 (HER2) overexpression. Consistent with the associated poor clinicopathologic features, patients with MAGE-A3/6-expressing tumors had a worse disease-specific survival as compared with patients with MAGE-A3/6 tumors. The frequent expression of MAGE-A3/6 in tumors of patients with primary HR BC, who have, for a large part, limited therapeutic options, encourages the selection of BC patients bearing MAGE-A3/6-expressing tumors for targeted immunotherapy.


Subject(s)
Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Breast Neoplasms/therapy , Carcinoma/diagnosis , Immunotherapy/methods , Neoplasm Proteins/metabolism , Antigens, Neoplasm/genetics , Breast Neoplasms/immunology , Breast Neoplasms/mortality , Carcinoma/immunology , Carcinoma/mortality , Female , Gene Expression Regulation, Neoplastic , Humans , Molecular Targeted Therapy , Neoplasm Proteins/genetics , Neoplasm Staging , Prognosis , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Survival Analysis
12.
Cancer Res ; 74(1): 68-80, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24197135

ABSTRACT

Beyond cancer-cell intrinsic factors, the immune status of the host has a prognostic impact on patients with cancer and influences the effects of conventional chemotherapies. Metastatic melanoma is intrinsically immunogenic, thereby facilitating the search for immune biomarkers of clinical responses to cytotoxic agents. Here, we show that a multi-tyrosine kinase inhibitor, sorafenib, upregulates interleukin (IL)-15Rα in vitro and in vivo in patients with melanoma, and in conjunction with natural killer (NK) group 2D (NKG2D) ligands, contributes to the Th1 polarization and accumulation of peripheral CD4(+)NKG2D(+) T cells. Hence, the increase of blood CD4(+)NKG2D(+) T cells after two cycles of sorafenib (combined with temozolomide) was associated with prolonged survival in a prospective phase I/II trial enrolling 63 patients with metastatic melanoma who did not receive vemurafenib nor immune checkpoint-blocking antibodies. In contrast, in metastatic melanoma patients treated with classical treatment modalities, this CD4(+)NKG2D(+) subset failed to correlate with prognosis. These findings indicate that sorafenib may be used as an "adjuvant" molecule capable of inducing or restoring IL-15Rα/IL-15 in tumors expressing MHC class I-related chain A/B (MICA/B) and on circulating monocytes of responding patients, hereby contributing to the bioactivity of NKG2D(+) Th1 cells.


Subject(s)
Interleukin-15 Receptor alpha Subunit/immunology , Melanoma/drug therapy , Melanoma/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Th1 Cells/immunology , Adult , Aged , CD4 Antigens/immunology , Cell Growth Processes/immunology , Female , Humans , Interleukin-15/immunology , Male , Melanoma/blood , Middle Aged , Niacinamide/therapeutic use , Sorafenib , Th1 Cells/drug effects , Young Adult
13.
J Immunol ; 191(9): 4619-27, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24068664

ABSTRACT

FOXP3(+) regulatory T cells (Tregs) are critical regulators of self-tolerance and immune homeostasis. In mice and humans, two subsets of FOXP3(+) Tregs have been defined based on their differential expression of Helios, a transcription factor of the Ikaros family. Whereas the origin, specificity, and differential function of the two subsets are as yet a source of controversy, their characterization thus far has been limited by the absence of surface markers to distinguish them. In this article, we show that human memory Helios(+) and Helios(-) Tregs are phenotypically distinct and can be separated ex vivo based on their differential expression of IL-1RI, which is restricted to Helios(-) Tregs, in combination with CCR7. The two populations isolated using this strategy are distinct with respect to the expression of other Ikaros family members. Namely, whereas Eos, which has been reported to mediate FOXP3-dependent gene silencing, is expressed in Helios(+) Tregs, Aiolos, which is involved in the differentiation of TH17 and induced Tregs, is instead expressed in Helios(-) Tregs. In addition, whereas both subsets are suppressive ex vivo, Helios(-) Tregs display increased suppressive capacity in comparison to Helios(+) Tregs, but respond to IL-1ß by downregulating their suppressive activity. Together, these data support the concept that human Helios(-) memory Tregs encompass induced Tregs that can readily respond to changes in the environment by modulating their suppressive capacity.


Subject(s)
Ikaros Transcription Factor/metabolism , Interleukin-1beta/metabolism , Receptors, Interleukin-1 Type I/metabolism , T-Lymphocytes, Regulatory/metabolism , Down-Regulation , Forkhead Transcription Factors/metabolism , Humans , Immunologic Memory , Interleukin-10/metabolism , Interleukin-17/metabolism , Receptors, CCR7/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology
15.
Cancer Immunol Res ; 1(5): 303-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24777968

ABSTRACT

Whereas tumor infiltration by T effectors is generally associated with a more favorable prognosis, the accumulation of CD4(+) regulatory T cells (Treg) within tumors is instead often associated with poor disease outcome. Because approaches to improve antitumor immunity aim, on one hand, at expanding tumor antigen-specific T cells and, on the other, at eliminating or inactivating Treg, an outstanding question is whether, and to what extent, tumor antigen-specific CD4(+) T effectors present at tumor sites overlap with tumor-associated Treg. Here, we used MHC class II/peptide tetramers incorporating an immunodominant peptide from the human tumor-specific antigen NY-ESO-1 to assess antigen-specific CD4(+) T cells among conventional CD4(+) T effectors and Treg at sites of ovarian cancer. We found that, in patients who spontaneously respond to the antigen, the frequency of NY-ESO-1 tetramer(+) cells detected ex vivo was highly enriched in tumors as compared with the periphery. At tumor sites, NY-ESO-1 tetramer(+) cells were detected concomitantly with high proportions of Treg but were distinct from the latter and displayed characteristics of TH1 effectors. Thus, even in the presence of high proportions of Treg, tumor antigen-specific CD4(+) T cells can accumulate in ovarian tumors and maintain an effector phenotype.


Subject(s)
Antigens, Neoplasm/immunology , Membrane Proteins/immunology , Ovarian Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Female , Humans
16.
Haematologica ; 98(2): 316-22, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22875619

ABSTRACT

Generation of tumor-antigen specific CD4(+) T-helper (T(H)) lines through in vitro priming is of interest for adoptive cell therapy of cancer, but the development of this approach has been limited by the lack of appropriate tools to identify and isolate low frequency tumor antigen-specific CD4(+) T cells. Here, we have used recently developed MHC class II/peptide tetramers incorporating an immunodominant peptide from NY-ESO-1 (ESO), a tumor antigen frequently expressed in different human solid and hematologic cancers, to implement an in vitro priming platform allowing the generation of ESO-specific T(H) lines. We isolated phenotypically defined CD4(+) T-cell subpopulations from circulating lymphocytes of DR52b(+) healthy donors by flow cytometry cell sorting and stimulated them in vitro with peptide ESO(119-143), autologous APC and IL-2. We assessed the frequency of ESO-specific cells in the cultures by staining with DR52b/ESO(119-143) tetramers (ESO-tetramers) and TCR repertoire of ESO-tetramer(+) cells by co-staining with TCR variable ß chain (BV) specific antibodies. We isolated ESO-tetramer(+) cells by flow cytometry cell sorting and expanded them with PHA, APC and IL-2 to generate ESO-specific T(H) lines. We characterized the lines for antigen recognition, by stimulation with ESO peptide or recombinant protein, cytokine production, by intracellular staining using specific antibodies, and alloreactivity, by stimulation with allo-APC. Using this approach, we could consistently generate ESO-tetramer(+) T(H) lines from conventional CD4(+)CD25(-) naïve and central memory populations, but not from effector memory populations or CD4(+)CD25(+) Treg. In vitro primed T(H) lines recognized ESO with affinities comparable to ESO-tetramer(+) cells from patients immunized with an ESO vaccine and used a similar TCR repertoire. In this study, using MHC class II/ESO tetramers, we have implemented an in vitro priming platform allowing the generation of ESO-monospecific polyclonal T(H) lines from non-immune individuals. This is an approach that is of potential interest for adoptive cell therapy of patients bearing ESO-expressing cancers.


Subject(s)
Antigens, Neoplasm/immunology , Histocompatibility Antigens Class II/immunology , Immunotherapy, Adoptive , Membrane Proteins/immunology , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes, Helper-Inducer/immunology , Antigen Presentation/immunology , Antigens, Neoplasm/metabolism , Cell Line , Cytokines/metabolism , Dendritic Cells/immunology , HLA-DR Serological Subtypes/immunology , HLA-DR Serological Subtypes/metabolism , Histocompatibility Antigens Class II/metabolism , Humans , Immunologic Memory , Membrane Proteins/metabolism , Protein Binding , Protein Multimerization , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocytes, Regulatory/immunology
17.
Cancer Res ; 72(17): 4351-60, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22798340

ABSTRACT

Antitumor type I T-cell responses involving IFN-γ production are critical to control cancer, but the efficacy of this response is limited by a variety of immunosuppressive mechanisms that promote tumoral immune escape. One critical mechanism involves the accumulation of FOXP3(+) T regulatory cells (Treg), a class of suppressive T cells that prevent excessive tissue destruction caused by unchecked immune responses. Recent studies have revealed that FOXP3(+) Treg include distinct subsets specifically controlling over the corresponding effector subset. In particular, CXCR3(+) Treg have been described as a subset specialized in the control of type I T-cell responses in vivo. Here, we show that CXCR3(+) Treg are highly enriched in human ovarian carcinomas, particularly in solid tumor masses, where they represent the majority of Treg. Tumor-associated CXCR3(+.) Treg coexpress T-bet but do not secrete IFN-γ ex vivo and suppress proliferation and IFN-γ secretion of T effectors. In addition, they coexpress Helios, suggesting that they originate from natural Treg. Finally, we show that the proportion of CXCR3(+) Treg at tumor sites is directly correlated with that of CXCR3(+) T effectors, consistent with expression of CXCR3 ligands. Together, our findings support the concept that natural CXCR3(+) T-bet(+) Treg selectively accumulate in ovarian tumors to control type I T-cell responses, resulting in the collateral limitation of efficient antitumor immunity.


Subject(s)
Carcinoma/immunology , Ovarian Neoplasms/immunology , Receptors, CXCR3/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Carcinoma/metabolism , Cell Line, Tumor , Female , Humans , Ikaros Transcription Factor/genetics , Ikaros Transcription Factor/metabolism , Interferon-gamma/metabolism , Ligands , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Ovarian Neoplasms/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism
19.
Cancer Res ; 72(5): 1059-63, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22253231

ABSTRACT

The role of T(H)17 cells in cancer is being investigated, but the existence of tumor antigen-specific T(H)17 cells has yet to be ascertained. Here, we report the first description of a spontaneous T(H)17 (IL-17(+)) response to the important tumor antigen MAGE-A3, which occurred concurrently with a T(H)1 (IFN-γ(+)) response in a lung cancer patient. MAGE-A3-specific interleukin (IL)-17(+) T cells were mainly CCR7(+) central memory T cells, whereas IFN-γ(+) cells were enriched for CCR7(-) effector memory T cells. An assessment of the fine specificity of antigen recognition by these T cells indicated that the CCR6(+)CCR4(+) and CCR6(+)CXCR3(+) fractions contained the same T(H)17/T(H)1 population at early and late differentiation stages, respectively, whereas the CCR6(-)CXCR3(+) fraction contained a distinct T(H)1 population. These findings are important because they suggest a differentiation model in which tumor antigen-specific CD4(+) T cells that are primed under T(H)17 polarizing conditions will progressively convert into IFN-γ-secreting cells in vivo as they differentiate into effector T cells that can effectively attack tumors.


Subject(s)
Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Interferon-gamma/metabolism , Lung Neoplasms/immunology , Neoplasm Proteins/immunology , Th17 Cells/immunology , Epitopes , Humans , Immunologic Memory , Interleukin-17/metabolism , Th1 Cells/immunology
20.
J Immunol ; 187(10): 5196-202, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21998454

ABSTRACT

IL-17-producing CD4(+) Th (Th17) cells are a unique subset of proinflammatory cells expressing the retinoic acid-related orphan receptor γt and associated with different forms of inflammatory autoimmune pathologies. The development of Th17 cells, mediated by TGF-ß and IL-1, is closely related to that of FOXP3(+) suppressor/regulatory T cells (Treg). In this study, we report that ex vivo expression of IL-1RI in human circulating CD4(+) T cells identifies a subpopulation of FOXP3(+) Treg that coexpress retinoic acid-related orphan receptor γt, secrete IL-17, and are highly enriched among CCR7(+) central memory cells. Consistent with the concept that IL-1RI expression in Treg identifies a subpopulation at an early stage of differentiation, we show that, in Th17 populations differentiated in vitro from natural naive FOXP3(+) Treg, IL-1RI(+) IL-17-secreting cells are central memory cells, whereas IL-1RI(-) cells secreting IL-17 are effector memory cells. Together with the absence of detectable IL-1RI and IL-17 expression in resting naive CD4(+) T cells, these data identify circulating CCR7(+) Treg expressing IL-1RI ex vivo as early intermediates along an IL-1-controlled differentiation pathway leading from naive FOXP3(+) Treg to Th17 effectors. We further show that, whereas IL-1RI(+) central memory Treg respond to stimulation in the presence of IL-1 by generating IL-17-secreting effectors, a significant fraction of them maintain FOXP3 expression, consistent with an important role of this population in maintaining the Treg/Th17 memory pool in vivo.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Forkhead Transcription Factors/biosynthesis , Receptors, Interleukin-1/biosynthesis , Th17 Cells/immunology , Humans , Immunologic Memory , Interleukin-17/metabolism , T-Lymphocytes, Regulatory/classification , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/cytology , Th17 Cells/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...