Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 165(2): 276-87, 2016 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-27058662

RESUMO

Anticancer immune responses can be considered a desirable form of autoimmunity that may be profoundly shaped by the microbiome. Here, we discuss evidence for the microbiome's influence on anti-tumor immunosurveillance, including those that are indirect and can act at a distance, and we put forward hypotheses regarding mechanisms of how these effects are implemented. These may involve cross-reactivity between microbial and tumor antigens shaping T cell repertoires and/or microbial products stimulating pattern recognition receptors that influence the type and intensity of immune responses. Understanding how the microbiome impacts natural cancer immunosurveillance as well as treatment-induced immune responses will pave the way for more effective therapies and prophylactics.


Assuntos
Terapia Biológica , Microbiota , Neoplasias/imunologia , Neoplasias/terapia , Animais , Disbiose , Humanos , Hipótese da Higiene , Monitorização Imunológica
2.
Immunity ; 53(4): 824-839.e10, 2020 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-33053331

RESUMO

CD8+ T cells within the tumor microenvironment (TME) are exposed to various signals that ultimately determine functional outcomes. Here, we examined the role of the co-activating receptor CD226 (DNAM-1) in CD8+ T cell function. The absence of CD226 expression identified a subset of dysfunctional CD8+ T cells present in peripheral blood of healthy individuals. These cells exhibited reduced LFA-1 activation, altered TCR signaling, and a distinct transcriptomic program upon stimulation. CD226neg CD8+ T cells accumulated in human and mouse tumors of diverse origin through an antigen-specific mechanism involving the transcriptional regulator Eomesodermin (Eomes). Despite similar expression of co-inhibitory receptors, CD8+ tumor-infiltrating lymphocyte failed to respond to anti-PD-1 in the absence of CD226. Immune checkpoint blockade efficacy was hampered in Cd226-/- mice. Anti-CD137 (4-1BB) agonists also stimulated Eomes-dependent CD226 loss that limited the anti-tumor efficacy of this treatment. Thus, CD226 loss restrains CD8+ T cell function and limits the efficacy of cancer immunotherapy.


Assuntos
Antígenos de Diferenciação de Linfócitos T/imunologia , Linfócitos T CD8-Positivos/imunologia , Neoplasias/imunologia , Proteínas com Domínio T/imunologia , Animais , Humanos , Inibidores de Checkpoint Imunológico/imunologia , Imunoterapia/métodos , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/terapia , Receptor de Morte Celular Programada 1/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Transdução de Sinais/imunologia , Transcriptoma/imunologia , Microambiente Tumoral/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia
3.
Eur J Immunol ; 45(2): 474-9, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25446972

RESUMO

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.


Assuntos
Fatores de Transcrição Forkhead/genética , Fator de Transcrição Ikaros/genética , L-Aminoácido Oxidase/genética , Linfócitos T Reguladores/metabolismo , Células Th17/metabolismo , Comunicação Celular , Diferenciação Celular , Fatores de Transcrição Forkhead/imunologia , Regulação da Expressão Gênica/imunologia , Humanos , Fator de Transcrição Ikaros/imunologia , Tolerância Imunológica , Interleucina-17/genética , Interleucina-17/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Ionomicina/farmacologia , L-Aminoácido Oxidase/imunologia , Ativação Linfocitária , Transdução de Sinais , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Acetato de Tetradecanoilforbol/farmacologia , Células Th17/citologia , Células Th17/efeitos dos fármacos , Células Th17/imunologia , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/imunologia
4.
Bull Acad Natl Med ; 200(1): 67-79, 2016 Jan.
Artigo em Inglês, Francês | MEDLINE | ID: mdl-29889414

RESUMO

Recent advances in cancer immunotherapy by immune checkpoint blockade are emphasizing the importance of Tcell responses to tumors in disease control. In addition, the development of next generation DNA sequencing technologies combined to reverse immunology approaches facilitate the appreciation of the involvement of T cell responses to neoantigens, which result from tumor-specific genomic alterations, in the immune response to tumors. Neoantigens extend the categories of previously characterized tumor antigens and, together with the latter, offer opportunities to target large proportions of tumors by passive and active specific immunotherapy approaches.


Assuntos
Antígenos de Neoplasias/imunologia , Imunoterapia/métodos , Neoplasias/terapia , Linfócitos T/imunologia , Análise Mutacional de DNA , Humanos , Neoplasias/imunologia
5.
J Immunol ; 191(9): 4619-27, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24068664

RESUMO

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.


Assuntos
Fator de Transcrição Ikaros/metabolismo , Interleucina-1beta/metabolismo , Receptores Tipo I de Interleucina-1/metabolismo , Linfócitos T Reguladores/metabolismo , Regulação para Baixo , Fatores de Transcrição Forkhead/metabolismo , Humanos , Memória Imunológica , Interleucina-10/metabolismo , Interleucina-17/metabolismo , Receptores CCR7/metabolismo , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia
6.
J Immunother Cancer ; 12(7)2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-39009451

RESUMO

BACKGROUND: Cervical cancer's lymphatic spread primarily begins from the sentinel lymph nodes (SLNs), underlining their pivotal role in disease metastasis. However, these nodes' immune gene expression profiles and immunoregulation mechanisms have yet to be explored. METHODS: Our study aimed to elucidate the immune cell populations and their roles in the immune gene expression profile of negative SLNs compared with positive SLNs and non-SLNs using Nanostring RNA seq analysis. We performed a principal component analysis on the log2 normalized expression of 685 endogenous genes in the nCounter PanCancer Immune Profiling Panel, followed by an assessment of the differential expression of genes and immune cell type abundance. RESULTS: We found significant variations in gene expression among the groups, with negative SLNs displaying overexpression of genes related to tumor-infiltrating immune cells, specifically innate cell populations. They also demonstrated the upregulation of genes involved in antigen presentation and T-cell priming. In contrast, positive SLNs were enriched in regulatory networks, suggesting their potential role in immune evasion. A comparison of negative SLNs and non-SLNs revealed increased innate and adaptive immune cell types, underscoring the ongoing T cell response to tumor antigens. CONCLUSION: Our findings underscore a specific immunogenetic phenotype profile in negative SLNs, emphasizing their crucial role in the initial anticancer response, immunosurveillance, and the propagation of immune tolerance from the primary cervical tumor. These results highlight the potential of SLNs as a novel target for immunotherapy strategies and underscore the importance of new imaging methods for accurately identifying SLN status without removal. Future investigations are needed to understand further the immunological interplay within SLNs and their influence on cervical cancer progression.


Assuntos
Linfonodo Sentinela , Neoplasias do Colo do Útero , Humanos , Feminino , Neoplasias do Colo do Útero/imunologia , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/patologia , Linfonodo Sentinela/patologia , Linfonodo Sentinela/imunologia , Imunogenética/métodos , Pessoa de Meia-Idade , Metástase Linfática , Biópsia de Linfonodo Sentinela/métodos
7.
Haematologica ; 98(2): 316-22, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22875619

RESUMO

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.


Assuntos
Antígenos de Neoplasias/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Imunoterapia Adotiva , Proteínas de Membrana/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Linfócitos T Auxiliares-Indutores/imunologia , Apresentação de Antígeno/imunologia , Antígenos de Neoplasias/metabolismo , Linhagem Celular , Citocinas/metabolismo , Células Dendríticas/imunologia , Subtipos Sorológicos de HLA-DR/imunologia , Subtipos Sorológicos de HLA-DR/metabolismo , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Memória Imunológica , Proteínas de Membrana/metabolismo , Ligação Proteica , Multimerização Proteica , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Linfócitos T Reguladores/imunologia
8.
J Immunol ; 187(10): 5196-202, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21998454

RESUMO

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.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Diferenciação Celular/imunologia , Fatores de Transcrição Forkhead/biossíntese , Receptores de Interleucina-1/biossíntese , Células Th17/imunologia , Humanos , Memória Imunológica , Interleucina-17/metabolismo , Linfócitos T Reguladores/classificação , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Células Th17/citologia , Células Th17/metabolismo , Fatores de Tempo
9.
Proc Natl Acad Sci U S A ; 107(45): 19402-7, 2010 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-20962281

RESUMO

RORγt(+) T(H)17 cells are a proinflammatory CD4(+) T-cell population associated with autoimmune tissue injury. In mice, priming of T(H)17 requires TGF-ß, which alone directs the priming of FOXP3(+) regulatory T cells (Treg), in association with inflammatory cytokines. Priming of human T(H)17 cells from conventional naive CD4(+) T cells under similar conditions, however, has proved difficult to achieve. Here, we report that differentiation of human T(H)17 cells preferentially occurs from FOXP3(+) naive Treg (NTreg) in the presence of IL-2 and IL-1ß and is increased by IL-23 and TGF-ß. IL-1ß-mediated differentiation correlated with IL-1RI expression in stimulated NTreg and was accompanied by induction of RORγt along with down-regulation of FOXP3. IL-17-secreting cells in NTreg cultures cosecreted TNF-α and IL-2 and contained distinct subpopulations cosecreting or not cosecreting IFN-γ and other T(H)17-associated cytokines. Polarized NTreg contained significant subpopulations of CCR6-expressing cells that were highly enriched in IL-17-secreting cells. Finally, analysis of CCR6 expression with respect to that of IL-1RI identified distinct IL-17-secreting subpopulations that had maintained or lost their suppressive functions. Together our results support the concept that priming of human T(H)17 from naive CD4(+) T cells preferentially takes place from FOXP3(+) Treg precursors in the presence of lineage-specific polarizing factors.


Assuntos
Diferenciação Celular/imunologia , Linhagem da Célula , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares , Linfócitos T Reguladores/citologia , Células Th17/citologia , Fatores de Transcrição Forkhead , Humanos , Interleucina-17/metabolismo , Interleucinas/farmacologia , Fator de Crescimento Transformador beta/farmacologia
10.
Proc Natl Acad Sci U S A ; 107(16): 7437-42, 2010 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-20368442

RESUMO

MHC-peptide tetramers have become essential tools for T-cell analysis, but few MHC class II tetramers incorporating peptides from human tumor and self-antigens have been developed. Among limiting factors are the high polymorphism of class II molecules and the low binding capacity of the peptides. Here, we report the generation of molecularly defined tetramers using His-tagged peptides and isolation of folded MHC/peptide monomers by affinity purification. Using this strategy we generated tetramers of DR52b (DRB3*0202), an allele expressed by approximately half of Caucasians, incorporating an epitope from the tumor antigen NY-ESO-1. Molecularly defined tetramers avidly and stably bound to specific CD4(+) T cells with negligible background on nonspecific cells. Using molecularly defined DR52b/NY-ESO-1 tetramers, we could demonstrate that in DR52b(+) cancer patients immunized with a recombinant NY-ESO-1 vaccine, vaccine-induced tetramer-positive cells represent ex vivo in average 1:5,000 circulating CD4(+) T cells, include central and transitional memory polyfunctional populations, and do not include CD4(+)CD25(+)CD127(-) regulatory T cells. This approach may significantly accelerate the development of reliable MHC class II tetramers to monitor immune responses to tumor and self-antigens.


Assuntos
Antígenos de Neoplasias/biossíntese , Linfócitos T CD4-Positivos/metabolismo , Antígenos de Histocompatibilidade Classe II/imunologia , Proteínas de Membrana/biossíntese , Neoplasias/imunologia , Peptídeos/química , Alelos , Antígenos/química , Epitopos/química , Citometria de Fluxo/métodos , Regulação Neoplásica da Expressão Gênica , Antígenos HLA-DR/química , Cadeias HLA-DRB3 , Humanos , Subunidade alfa de Receptor de Interleucina-2/biossíntese , Neoplasias/sangue , Fenótipo , Proteínas Recombinantes/química
11.
Sci Immunol ; 8(84): eadg8841, 2023 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-37289857

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos , Neoplasias do Colo , Humanos , Prognóstico , Integrinas
12.
Front Oncol ; 13: 1269166, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38074683

RESUMO

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.

13.
Proc Natl Acad Sci U S A ; 106(21): 8635-40, 2009 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-19439651

RESUMO

Recent studies have suggested a close relationship between CD4(+)FOXP3(+) regulatory T cells (Tregs) and proinflammatory IL-17-producing T helper cells (T(H)17) expressing the lineage-specific transcription factor RORgamma t. We report here the unexpected finding that human memory Tregs secrete IL-17 ex vivo and constitutively express RORgamma t. IL-17-secreting Tregs share some phenotypic and functional features with conventional T(H)17 cells, expressing high levels of CCR4 and CCR6 and low levels of CXCR3. However, unlike conventional T(H)17 cells, they express low levels of CD161 and mostly fail to cosecrete IL-22 and TNF-alpha ex vivo. Ex vivo secretion of IL-17 and constitutive expression of RORgamma t by human memory Tregs suggest that, in addition to their well-known suppressive functions, these cells likely play additional, as yet undescribed, proinflammatory functions.


Assuntos
Linhagem da Célula/imunologia , Fatores de Transcrição Forkhead/imunologia , Memória Imunológica/imunologia , Interleucina-17/metabolismo , Receptores do Ácido Retinoico/imunologia , Receptores dos Hormônios Tireóideos/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Reguladores/imunologia , Humanos , Interleucina-17/imunologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares , Fenótipo , Linfócitos T Reguladores/metabolismo
14.
Cancers (Basel) ; 14(15)2022 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-35954341

RESUMO

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.

16.
Front Oncol ; 11: 662236, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33968769

RESUMO

Radiation-induced immune effects have been extensively deciphered over the last few years, leading to the concept of the dual immune effect of radiotherapy with both immunostimulatory and immunosuppressive effects. This explains why radiotherapy alone is not able to drive a strong anti-tumor immune response in most cases, hence underlining the rationale for combining both radiotherapy and immunotherapy. This association has generated considerable interest and hundreds of trials are currently ongoing to assess such an association in oncology. However, while some trials have provided unprecedented results or shown much promise, many hopes have been dashed. Questions remain, therefore, as to how to optimize the combination of these treatment modalities. This narrative review aims at revisiting the old, well-established concepts of radiotherapy relating to dose, fractionation, target volumes and organs at risk in the era of immunotherapy. We then propose potential innovative approaches to be further assessed when considering a radio-immunotherapy association, especially in the field of non-small-cell lung cancer (NSCLC). We finally propose a framework to optimize the association, with pragmatic approaches depending on the stage of the disease.

17.
Oncoimmunology ; 10(1): 1939518, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34721945

RESUMO

γδ T lymphocytes diverge from conventional T CD8 lymphocytes for ontogeny, homing, and antigen specificity, but whether their differentiation in tumors also deviates was unknown. Using innovative analyses of our original and ~150 published single-cell RNA sequencing datasets validated by phenotyping of human tumors and murine models, here we present the first high-resolution view of human γδ T cell differentiation in cancer. While γδ T lymphocytes prominently encompass TCRVγ9 cells more differentiated than T CD8 in healthy donor's blood, a different scenario is unveiled in tumors. Solid tumors and lymphomas are infiltrated by a majority of TCRVγnon9 γδ T cells which are quantitatively correlated and remarkably aligned with T CD8 for differentiation, exhaustion, gene expression profile, and response to immune checkpoint therapy. This cancer-wide association is critical for developing cancer immunotherapies.


Assuntos
Neoplasias , Transcriptoma , Animais , Linfócitos T CD8-Positivos , Diferenciação Celular , Humanos , Linfócitos do Interstício Tumoral , Camundongos , Neoplasias/genética , Receptores de Antígenos de Linfócitos T gama-delta/genética , Subpopulações de Linfócitos T
18.
Clin Cancer Res ; 27(4): 1037-1047, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33272982

RESUMO

PURPOSE: TNF blockers can be used to manage gastrointestinal inflammatory side effects following nivolumab and/or ipilimumab treatment in patients with advanced melanoma. Our preclinical data showed that anti-TNF could promote the efficacy of immune checkpoint inhibitors. PATIENTS AND METHODS: TICIMEL (NTC03293784) is an open-label, two-arm phase Ib clinical trial. Fourteen patients with advanced and/or metastatic melanoma (stage IIIc/IV) were enrolled. Patients were treated with nivolumab (1 mg/kg) and ipilimumab (3 mg/kg) combined to infliximab (5 mg/kg, N = 6) or certolizumab (400/200 mg, N = 8). The primary endpoint was safety and the secondary endpoint was antitumor activity. Adverse events (AEs) were graded according to the NCI Common Terminology Criteria for Adverse Events and response was assessed following RECIST 1.1. RESULTS: Only one dose-limiting toxicity was observed in the infliximab cohort. The two different combinations were found to be safe. We observed lower treatment-related AEs with infliximab as compared with certolizumab. In the certolizumab cohort, one patient was not evaluable for response. In this cohort, four of eight patients exhibited hepatobiliary disorders and seven of seven evaluable patients achieved objective response including four complete responses (CRs) and three partial responses (PRs). In the infliximab cohort, we observed one CR, two PRs, and three progressive diseases. Signs of activation and maturation of systemic T-cell responses were seen in patients from both cohorts. CONCLUSIONS: Our results show that both combinations are safe in human and provide clinical and biological activities. The high response rate in the certolizumab-treated patient cohort deserves further investigations.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Melanoma/tratamento farmacológico , Neoplasias Cutâneas/tratamento farmacológico , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Certolizumab Pegol/administração & dosagem , Certolizumab Pegol/efeitos adversos , Feminino , Seguimentos , Humanos , Infliximab/administração & dosagem , Infliximab/efeitos adversos , Ipilimumab/administração & dosagem , Ipilimumab/efeitos adversos , Masculino , Melanoma/diagnóstico , Melanoma/mortalidade , Melanoma/secundário , Pessoa de Meia-Idade , Nivolumabe/administração & dosagem , Nivolumabe/efeitos adversos , Intervalo Livre de Progressão , Neoplasias Cutâneas/diagnóstico , Neoplasias Cutâneas/mortalidade , Neoplasias Cutâneas/patologia , Resultado do Tratamento , Fator de Necrose Tumoral alfa/antagonistas & inibidores
19.
JCI Insight ; 6(2)2021 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-33332284

RESUMO

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.


Assuntos
Linfócitos do Interstício Tumoral/imunologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Linfócitos T Auxiliares-Indutores/imunologia , Antígenos de Neoplasias/imunologia , Apirase/imunologia , Linfócitos T CD8-Positivos/imunologia , Feminino , Expressão Gênica , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/imunologia , Humanos , Tolerância Imunológica/genética , Imunidade Celular/genética , Técnicas In Vitro , Ativação Linfocitária/genética , Cooperação Linfocítica/genética , Masculino , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/imunologia , Receptor de Morte Celular Programada 1/imunologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Evasão Tumoral/genética , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA