Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Front Oncol ; 13: 1269166, 2023.
Article in English | MEDLINE | ID: mdl-38074683

ABSTRACT

Background: While much progress has been accomplished in the understanding of radiation-induced immune effects in tumors, little is known regarding the mechanisms involved at the tumor draining lymph node (TDLN) level. The objective of this retrospective study was to assess the immune and biological changes arising in non-involved TDLNs upon node sparing concurrent chemoradiotherapy (CRT) of non-small cell lung cancer (NSCLC) tumors. Methods: Patients with proven localized (cN0M0) NSCLC, treated by radical surgery plus lymph node dissection with (CRT+) or without (CRT-) neoadjuvant chemoradiotherapy, whereby radiotherapy was targeted on the primary tumor with no significant incidental irradiation of the non-involved TDLN station (stations XI), were identified. Bulk RNA sequencing of TDLNs was performed and data were analyzed based on differential gene expression (DGE) and gene sets enrichment. Results: Sixteen patients were included and 25 TDLNs were analyzed: 6 patients in the CRT+ group (12 samples) and 10 patients in the CRT- group (13 samples). Overall, 1001 genes were differentially expressed between the two groups (CRT+ and CRT-). Analysis with g-profiler revealed that gene sets associated with antitumor immune response, inflammatory response, hypoxia, angiogenesis, epithelial mesenchymal transition and extra-cellular matrix remodeling were enriched in the CRT+ group, whereas only gene sets associated with B cells and B-cell receptor signaling were enriched in the CRT- group. Unsupervised dimensionality reduction identified two clusters of TDLNs from CRT+ patients, of which one cluster (cluster 1) exhibited higher expression of pathways identified as enriched in the overall CRT+ group in comparison to the CRT- group. In CRT+ cluster 1, 3 out of 3 patients had pathological complete response (pCR) or major pathological response (MPR) to neoadjuvant CRT, whereas only 1 out of 3 patients in the other CRT+ cluster (cluster 2) experienced MPR and none exhibited pCR. Conclusion: Neoadjuvant node sparing concurrent CRT of NSCLC patients is associated with distinct microenvironment and immunological patterns in non-involved TDLNs as compared to non-involved TDLNs from patients with non-irradiated tumors. Our data are in line with studies showing superiority of lymph node sparing irradiation of the primary tumor in the induction of antitumor immunity.

2.
Sci Immunol ; 8(84): eadg8841, 2023 06 02.
Article in English | MEDLINE | ID: mdl-37289857

ABSTRACT

Despite the high prognostic value of immune infiltrates in colorectal cancer (CRC), metastatic disease remains resistant to immunotherapy by immune checkpoint blockade (ICB). Here, we show, in metastatic CRC preclinical models, that orthotopically implanted primary colon tumors exert a colon-specific antimetastatic effect on distant hepatic lesions. Enterotropic α4ß7 integrin-expressing neoantigen-specific CD8 T cells were key components of the antimetastatic effect. Accordingly, the presence of concomitant colon tumors improved control of liver lesions by anti-PD-L1 proof-of-concept immunotherapy and generated protective immune memory, whereas partial depletion of α4ß7+ cells abrogated control of metastases. Last, in patients with metastatic CRC, response to ICB was associated with expression of α4ß7 integrin in metastases and with circulating α4ß7+ CD8 T cells. Our findings identify a systemic cancer immunosurveillance role for gut-primed tumor-specific α4ß7+ CD8 T cells.


Subject(s)
CD8-Positive T-Lymphocytes , Colonic Neoplasms , Humans , Prognosis , Integrins
3.
Cancers (Basel) ; 14(15)2022 Jul 28.
Article in English | MEDLINE | ID: mdl-35954341

ABSTRACT

Tumor-infiltrating exhausted PD-1hiCD39+ tumor-antigen (Ag)-specific CD4 T cells contribute to the response to immune checkpoint blockade (ICB), but their circulating counterparts, which could represent accessible biomarkers, have not been assessed. Here, we analyzed circulating PD-1+CD39+ CD4 T cells and show that this population was present at higher proportions in cancer patients than in healthy individuals and was enriched in activated HLA-DR+ and ICOS+ and proliferating KI67+ cells, indicative of their involvement in ongoing immune responses. Among memory CD4 T cells, this population contained the lowest proportions of cells producing effector cytokines, suggesting they were exhausted. In patients with HPV-induced malignancies, the PD-1+CD39+ population contained high proportions of HPV Ag-specific T cells. In patients treated by ICB for HPV-induced tumors, the proportion of circulating PD-1+CD39+ CD4 T cells was predictive of the clinical response. Our results identify CD39 expression as a surrogate marker of circulating helper tumor-Ag-specific CD4 T cells.

4.
JCI Insight ; 6(2)2021 01 25.
Article in English | MEDLINE | ID: mdl-33332284

ABSTRACT

Tumor antigen-specific CD4 T cells accumulate at tumor sites, evoking their involvement in antitumor effector functions in situ. Contrary to CD8 cytotoxic T lymphocyte exhaustion, that of CD4 T cells remains poorly appreciated. Here, using phenotypic, transcriptomic, and functional approaches, we characterized CD4 T cell exhaustion in patients with head and neck, cervical, and ovarian cancer. We identified a CD4 tumor-infiltrating lymphocyte (TIL) population, defined by high PD-1 and CD39 expression, which contained high proportions of cytokine-producing cells, although the quantity of cytokines produced by these cells was low, evoking an exhausted state. Terminal exhaustion of CD4 TILs was instated regardless of TIM-3 expression, suggesting divergence with CD8 T cell exhaustion. scRNA-Seq and further phenotypic analyses uncovered similarities with the CD8 T cell exhaustion program. In particular, PD-1hiCD39+ CD4 TILs expressed the exhaustion transcription factor TOX and the chemokine CXCL13 and were tumor antigen specific. In vitro, PD-1 blockade enhanced CD4 TIL activation, as evidenced by increased CD154 expression and cytokine secretion, leading to improved dendritic cell maturation and consequently higher tumor-specific CD8 T cell proliferation. Our data identify exhausted CD4 TILs as players in responsiveness to immune checkpoint blockade.


Subject(s)
Lymphocytes, Tumor-Infiltrating/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes, Helper-Inducer/immunology , Antigens, Neoplasm/immunology , Apyrase/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Gene Expression , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/immunology , Humans , Immune Tolerance/genetics , Immunity, Cellular/genetics , In Vitro Techniques , Lymphocyte Activation/genetics , Lymphocyte Cooperation/genetics , Male , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Programmed Cell Death 1 Receptor/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Escape/genetics , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/immunology
5.
Science ; 369(6506): 936-942, 2020 08 21.
Article in English | MEDLINE | ID: mdl-32820119

ABSTRACT

Intestinal microbiota have been proposed to induce commensal-specific memory T cells that cross-react with tumor-associated antigens. We identified major histocompatibility complex (MHC) class I-binding epitopes in the tail length tape measure protein (TMP) of a prophage found in the genome of the bacteriophage Enterococcus hirae Mice bearing E. hirae harboring this prophage mounted a TMP-specific H-2Kb-restricted CD8+ T lymphocyte response upon immunotherapy with cyclophosphamide or anti-PD-1 antibodies. Administration of bacterial strains engineered to express the TMP epitope improved immunotherapy in mice. In renal and lung cancer patients, the presence of the enterococcal prophage in stools and expression of a TMP-cross-reactive antigen by tumors correlated with long-term benefit of PD-1 blockade therapy. In melanoma patients, T cell clones recognizing naturally processed cancer antigens that are cross-reactive with microbial peptides were detected.


Subject(s)
Antigens, Neoplasm/immunology , Bacteriophages/immunology , Enterococcus hirae/virology , Gastrointestinal Microbiome/immunology , Histocompatibility Antigens Class I/immunology , Immunotherapy/methods , Neoplasms/therapy , Viral Tail Proteins/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents, Alkylating/therapeutic use , CD8-Positive T-Lymphocytes/immunology , Cross Reactions , Cyclophosphamide/therapeutic use , Epitopes/immunology , Feces/virology , H-2 Antigens/immunology , Humans , Mice , Neoplasms/diet therapy , Neoplasms/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Viral Tail Proteins/therapeutic use
6.
Cancer Immunol Res ; 8(7): 869-882, 2020 07.
Article in English | MEDLINE | ID: mdl-32295784

ABSTRACT

Although understanding of T-cell exhaustion is widely based on mouse models, its analysis in patients with cancer could provide clues indicating tumor sensitivity to immune checkpoint blockade (ICB). Data suggest a role for costimulatory pathways, particularly CD28, in exhausted T-cell responsiveness to PD-1/PD-L1 blockade. Here, we used single-cell transcriptomic, phenotypic, and functional approaches to dissect the relation between CD8+ T-cell exhaustion, CD28 costimulation, and tumor specificity in head and neck, cervical, and ovarian cancers. We found that memory tumor-specific CD8+ T cells, but not bystander cells, sequentially express immune checkpoints once they infiltrate tumors, leading, in situ, to a functionally exhausted population. Exhausted T cells were nonetheless endowed with effector and tumor residency potential but exhibited loss of the costimulatory receptor CD28 in comparison with their circulating memory counterparts. Accordingly, PD-1 inhibition improved proliferation of circulating tumor-specific CD8+ T cells and reversed functional exhaustion of specific T cells at tumor sites. In agreement with their tumor specificity, high infiltration of tumors by exhausted cells was predictive of response to therapy and survival in ICB-treated patients with head and neck cancer. Our results showed that PD-1 blockade-mediated proliferation/reinvigoration of circulating memory T cells and local reversion of exhaustion occur concurrently to control tumors.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , CD28 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Neoplasms, Glandular and Epithelial/drug therapy , Neoplasms, Glandular and Epithelial/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , CD28 Antigens/metabolism , CD8-Positive T-Lymphocytes/drug effects , Cell Proliferation/physiology , Female , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Single-Cell Analysis/methods , Survival Rate , Transcriptome
7.
Cell Rep ; 26(1): 94-107.e7, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30605689

ABSTRACT

Despite the clinical success of blocking inhibitory immune checkpoint receptors such as programmed cell death-1 (PD-1) in cancer, the mechanisms controlling the expression of these receptors have not been fully elucidated. Here, we identify a post-transcriptional mechanism regulating PD-1 expression in T cells. Upon activation, the PDCD1 mRNA and ribonucleoprotein complexes coalesce into stress granules that require microtubules and the kinesin 1 molecular motor to proceed to translation. Hence, PD-1 expression is highly sensitive to microtubule or stress granule inhibitors targeting this pathway. Evidence from healthy donors and cancer patients reveals a common regulation for the translation of CTLA4, LAG3, TIM3, TIGIT, and BTLA but not of the stimulatory co-receptors OX40, GITR, and 4-1BB mRNAs. In patients, disproportionality analysis of immune-related adverse events for currently used microtubule drugs unveils a significantly higher risk of autoimmunity. Our findings reveal a fundamental mechanism of immunoregulation with great importance in cancer immunotherapy.


Subject(s)
Immunotherapy/methods , Microtubules/metabolism , T-Lymphocytes/immunology , Humans
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
9.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...