Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Semin Immunol ; 49: 101436, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-33288379

RESUMEN

T cells play a critical role in promoting tumor regression in both experimental models and humans. Yet, T cells that are chronically exposed to tumor antigen during cancer progression can become dysfunctional/exhausted and fail to induce tumor destruction. Such tumor-induced T cell dysfunction may occur via multiple mechanisms. In particular, immune checkpoint inhibitory receptors that are upregulated by tumor-infiltrating lymphocytes in many cancers limit T cell survival and function. Overcoming this inhibitory receptor-mediated T cell dysfunction has been a central focus of recent developments in cancer immunotherapy. Immunotherapies targeting inhibitory receptor pathways such as programmed cell death 1 (PD-1)/programmed death ligand 1 and cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), alone or in combination, confer significant clinical benefits in multiple tumor types. However, many patients with cancer do not respond to immune checkpoint blockade, and dual PD-1/CTLA-4 blockade may cause serious adverse events, which limits its indications. Targeting novel non-redundant inhibitory receptor pathways contributing to tumor-induced T cell dysfunction in the tumor microenvironment may prove efficacious and non-toxic. This review presents preclinical and clinical findings supporting the roles of two key pathways-T-cell immunoglobulin and mucin-domain containing-3 (TIM-3) and T cell immunoreceptor with Ig and ITIM domain (TIGIT)/CD226/CD96/CD112R-in cancer immunotherapy.


Asunto(s)
Receptores Coestimuladores e Inhibidores de Linfocitos T/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Animales , Biomarcadores de Tumor , Humanos , Proteínas de Punto de Control Inmunitario/metabolismo , Inmunoterapia , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Terapia Molecular Dirigida , Neoplasias/patología , Neoplasias/terapia , Transducción de Señal , Subgrupos de Linfocitos T/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
2.
PLoS Biol ; 12(4): e1001833, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24714093

RESUMEN

How the microbiota affects health and disease is a crucial question. In mice, gut Clostridium bacteria are potent inducers of colonic interleukin (IL)-10-producing Foxp3 regulatory T cells (Treg), which play key roles in the prevention of colitis and in systemic immunity. In humans, although gut microbiota dysbiosis is associated with immune disorders, the underlying mechanism remains unknown. In contrast with mice, the contribution of Foxp3 Treg in colitis prevention has been questioned, suggesting that other compensatory regulatory cells or mechanisms may exist. Here we addressed the regulatory role of the CD4CD8 T cells whose presence had been reported in the intestinal mucosa and blood. Using colonic lamina propria lymphocytes (LPL) and peripheral blood lymphocytes (PBL) from healthy individuals, and those with colon cancer and irritable bowel disease (IBD), we demonstrated that CD4CD8αα (DP8α) T lymphocytes expressed most of the regulatory markers and functions of Foxp3 Treg and secreted IL-10. Strikingly, DP8α LPL and PBL exhibited a highly skewed repertoire toward the recognition of Faecalibacterium prausnitzii, a major Clostridium species of the human gut microbiota, which is decreased in patients with IBD. Furthermore, the frequencies of DP8α PBL and colonic LPL were lower in patients with IBD than in healthy donors and in the healthy mucosa of patients with colon cancer, respectively. Moreover, PBL and LPL from most patients with active IBD failed to respond to F. prausnitzii in contrast to PBL and LPL from patients in remission and/or healthy donors. These data (i) uncover a Clostridium-specific IL-10-secreting Treg subset present in the human colonic LP and blood, (ii) identify F. prausnitzii as a major inducer of these Treg, (iii) argue that these cells contribute to the control or prevention of colitis, opening new diagnostic and therapeutic strategies for IBD, and (iv) provide new tools to address the systemic impact of both these Treg and the intestinal microbiota on the human immune homeostasis.


Asunto(s)
Clostridium/inmunología , Colitis Ulcerosa/inmunología , Enfermedad de Crohn/inmunología , Mucosa Intestinal/citología , Linfocitos T Reguladores/inmunología , Antígenos CD4/metabolismo , Antígenos CD8/metabolismo , Linfocitos T CD8-positivos/inmunología , Colon/inmunología , Colon/microbiología , Neoplasias del Colon/inmunología , Factores de Transcripción Forkhead/biosíntesis , Humanos , Interleucina-10/biosíntesis , Mucosa Intestinal/inmunología , Subgrupos de Linfocitos T/inmunología
3.
J Immunol ; 188(5): 2102-10, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22291187

RESUMEN

The uptake and long-term cross-presentation of tumor Ag long peptides (LP) by dendritic cells (DC) make them attractive cancer vaccine candidates. However, it remains to be established whether LP can prime long-lived tumor-reactive CTL and whether other cell types are able to cross-present them. Using HLA-A2 healthy donor and melanoma patient-derived PBMC, we studied the in vitro cross-priming potential of Melan-A 16-40 LP bearing the HLA-A2-restricted epitope 26-35 or its analog 26-35(A27L) and compared it to the priming capacity of the short analog. We then addressed LP priming capacity in vivo using HLA-A2 mice. We also studied LP cross-presentation by monocyte-derived DC, plasmacytoid DC, monocytes, and B cells. We showed that the modified LP gave rise to high and sustained cross-presentation by monocyte-derived DC. This led to cross priming in vitro and in vivo and to expansion of long-lived tumor-reactive cytotoxic T cells. In contrast, the LP containing the natural 26-35 epitope primed specific T cells poorly, despite its long-lived cross-presentation, and T cells primed against the short analog were short-lived. We further showed that LP cross-presentation is restricted to monocytes and conventional DC. These results document for the first time, to our knowledge, the strong immunogenicity of a human tumor Ag LP. Of note, they underscore that this property is critically dependent on sufficient HLA binding affinity and/or TCR ligand potency of the cross-presented epitope. We conclude that LP fulfilling this requirement should be used as tumor vaccines, together with DC maturating agents, especially the Melan-A 16-40(A27L) LP, for the treatment of HLA-A2(+) melanoma patients.


Asunto(s)
Neoplasias Colorrectales/inmunología , Reactividad Cruzada/inmunología , Epítopos de Linfocito T/metabolismo , Antígeno HLA-A2/metabolismo , Antígeno MART-1/metabolismo , Melanoma/inmunología , Fragmentos de Péptidos/metabolismo , Linfocitos T Citotóxicos/inmunología , Secuencia de Aminoácidos , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Vacunas contra el Cáncer/farmacología , Vacunas contra el Cáncer/uso terapéutico , Línea Celular Tumoral , Células Cultivadas , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Epítopos de Linfocito T/fisiología , Antígeno HLA-A2/fisiología , Humanos , Epítopos Inmunodominantes/metabolismo , Epítopos Inmunodominantes/fisiología , Activación de Linfocitos/inmunología , Antígeno MART-1/fisiología , Melanoma/patología , Melanoma/terapia , Ratones , Ratones Mutantes , Datos de Secuencia Molecular , Monocitos/inmunología , Monocitos/metabolismo , Fragmentos de Péptidos/fisiología , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/patología
4.
Nat Med ; 28(3): 545-556, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35228752

RESUMEN

Ample evidence indicates that the gut microbiome is a tumor-extrinsic factor associated with antitumor response to anti-programmed cell death protein-1 (PD-1) therapy, but inconsistencies exist between published microbial signatures associated with clinical outcomes. To resolve this, we evaluated a new melanoma cohort, along with four published datasets. Time-to-event analysis showed that baseline microbiota composition was optimally associated with clinical outcome at approximately 1 year after initiation of treatment. Meta-analysis and other bioinformatic analyses of the combined data show that bacteria associated with favorable response are confined within the Actinobacteria phylum and the Lachnospiraceae/Ruminococcaceae families of Firmicutes. Conversely, Gram-negative bacteria were associated with an inflammatory host intestinal gene signature, increased blood neutrophil-to-lymphocyte ratio, and unfavorable outcome. Two microbial signatures, enriched for Lachnospiraceae spp. and Streptococcaceae spp., were associated with favorable and unfavorable clinical response, respectively, and with distinct immune-related adverse effects. Despite between-cohort heterogeneity, optimized all-minus-one supervised learning algorithms trained on batch-corrected microbiome data consistently predicted outcomes to programmed cell death protein-1 therapy in all cohorts. Gut microbial communities (microbiotypes) with nonuniform geographical distribution were associated with favorable and unfavorable outcomes, contributing to discrepancies between cohorts. Our findings shed new light on the complex interaction between the gut microbiome and response to cancer immunotherapy, providing a roadmap for future studies.


Asunto(s)
Microbioma Gastrointestinal , Melanoma , Microbiota , Bacterias/genética , Microbioma Gastrointestinal/genética , Humanos , Inmunoterapia/efectos adversos , Melanoma/tratamiento farmacológico
5.
J Clin Invest ; 132(9)2022 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-35316223

RESUMEN

T cell immunoglobulin mucin domain-containing protein 3 (Tim-3) negatively regulates innate and adaptive immunity in cancer. To identify the mechanisms of Tim-3 in cancer immunity, we evaluated the effects of Tim-3 blockade in human and mouse melanoma. Here, we show that human programmed cell death 1-positive (PD-1+) Tim-3+CD8+ tumor-infiltrating lymphocytes (TILs) upregulate phosphatidylserine (PS), a receptor for Tim-3, and acquire cell surface myeloid markers from antigen-presenting cells (APCs) through transfer of membrane fragments called trogocytosis. Tim-3 blockade acted on Tim-3+ APCs in a PS-dependent fashion to disrupt the trogocytosis of activated tumor antigen-specific CD8+ T cells and PD-1+Tim-3+ CD8+ TILs isolated from patients with melanoma. Tim-3 and PD-1 blockades cooperated to disrupt trogocytosis of CD8+ TILs in 2 melanoma mouse models, decreasing tumor burden and prolonging survival. Deleting Tim-3 in dendritic cells but not in CD8+ T cells impeded the trogocytosis of CD8+ TILs in vivo. Trogocytosed CD8+ T cells presented tumor peptide-major histocompatibility complexes and became the target of fratricide T cell killing, which was reversed by Tim-3 blockade. Our findings have uncovered a mechanism Tim-3 uses to limit antitumor immunity.


Asunto(s)
Receptor 2 Celular del Virus de la Hepatitis A/inmunología , Melanoma , Animales , Linfocitos T CD8-positivos , Receptor 2 Celular del Virus de la Hepatitis A/genética , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Humanos , Linfocitos Infiltrantes de Tumor , Melanoma/patología , Ratones , Receptor de Muerte Celular Programada 1 , Trogocitosis
6.
Science ; 371(6529): 595-602, 2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33542131

RESUMEN

Anti-programmed cell death protein 1 (PD-1) therapy provides long-term clinical benefits to patients with advanced melanoma. The composition of the gut microbiota correlates with anti-PD-1 efficacy in preclinical models and cancer patients. To investigate whether resistance to anti-PD-1 can be overcome by changing the gut microbiota, this clinical trial evaluated the safety and efficacy of responder-derived fecal microbiota transplantation (FMT) together with anti-PD-1 in patients with PD-1-refractory melanoma. This combination was well tolerated, provided clinical benefit in 6 of 15 patients, and induced rapid and durable microbiota perturbation. Responders exhibited increased abundance of taxa that were previously shown to be associated with response to anti-PD-1, increased CD8+ T cell activation, and decreased frequency of interleukin-8-expressing myeloid cells. Responders had distinct proteomic and metabolomic signatures, and transkingdom network analyses confirmed that the gut microbiome regulated these changes. Collectively, our findings show that FMT and anti-PD-1 changed the gut microbiome and reprogrammed the tumor microenvironment to overcome resistance to anti-PD-1 in a subset of PD-1 advanced melanoma.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Resistencia a Antineoplásicos , Trasplante de Microbiota Fecal , Melanoma/terapia , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Neoplasias Cutáneas/terapia , Linfocitos T CD8-positivos/inmunología , Microbioma Gastrointestinal , Humanos , Interleucina-8/inmunología , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor/inmunología , Células Mieloides/inmunología , Microambiente Tumoral/inmunología
7.
Biol Blood Marrow Transplant ; 16(3): 430-4, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19883775

RESUMEN

Peripheral blood stem cell transplantation (PBSCT) is an alternative to bone marrow transplantation (BMT). Although CD4(+)CD25(+)CD127(lo) regulatory T cells (Tregs) have been shown to play important roles in the control of T cell reactivity, the Treg contents of both graft types have not been analyzed comparatively to date. We report herein that Treg frequencies are significantly reduced in PBSC compared to BM transplants. Furthermore, most Tregs from PBSC transplants are CD62L(lo), a phenotype reported to have poor suppressor activity. Both granulocyte-colony stimulating factor (G-CSF) administration and leukapheresis were found to contribute to the loss of CD62L(+) Tregs. Although higher T cell numbers are infused in PBSCT than in BMT, it is possible that the reduced Treg content of PBSC transplants may represent 1 factor contributing to the higher risk of GVHD reported after PBSCT.


Asunto(s)
Trasplante de Médula Ósea , Trasplante de Células Madre de Sangre Periférica , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/trasplante , Apoptosis/inmunología , Médula Ósea/efectos de los fármacos , Factores de Transcripción Forkhead/análisis , Enfermedad Injerto contra Huésped/inmunología , Factor Estimulante de Colonias de Granulocitos/farmacología , Humanos , Inmunofenotipificación , Subunidad alfa del Receptor de Interleucina-7/análisis , Selectina L/análisis , Leucaféresis , Antígenos Comunes de Leucocito/análisis , Recuento de Linfocitos , Factores de Riesgo , Subgrupos de Linfocitos T/química , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/trasplante , Linfocitos T Reguladores/química , Linfocitos T Reguladores/inmunología , Donantes de Tejidos
8.
J Immunother Cancer ; 8(2)2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32900861

RESUMEN

Tumors evade immune-mediated recognition through multiple mechanisms of immune escape. On chronic tumor antigen exposure, T cells become dysfunctional/exhausted and upregulate various checkpoint inhibitory receptors (IRs) that limit T cells' survival and function. During the last decade, immunotherapies targeting IRs such as programmed cell death receptor 1 (PD-1) and anticytotoxic T lymphocyte-associated antigen 4 (CTLA-4) have provided ample evidence of clinical benefits in many solid tumors. Beyond CTLA-4 and PD-1, multiple other IRs are also targeted with immune checkpoint blockade in the clinic. Specifically, T cell immunoreceptor with immunoglobulin and ITIM domain (TIGIT) is a promising new target for cancer immunotherapy. TIGIT is upregulated by immune cells, including activated T cells, natural killer cells, and regulatory T cells. TIGIT binds to two ligands, CD155 (PVR) and CD112 (PVRL2, nectin-2), that are expressed by tumor cells and antigen-presenting cells in the tumor microenvironment. There is now ample evidence that the TIGIT pathway regulates T cell-mediated and natural killer cell-mediated tumor recognition in vivo and in vitro. Dual PD-1/TIGIT blockade potently increases tumor antigen-specific CD8+ T cell expansion and function in vitro and promotes tumor rejection in mouse tumor models. These findings support development of ongoing clinical trials with dual PD-1/TIGIT blockade in patients with cancer.


Asunto(s)
Inmunoterapia/métodos , Neoplasias/terapia , Receptores Inmunológicos/metabolismo , Animales , Humanos , Ratones
9.
Clin Cancer Res ; 26(20): 5520-5533, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32591463

RESUMEN

PURPOSE: Natural killer (NK) cells play a critical role in tumor immunosurveillance. Multiple activating and inhibitory receptors (IR) regulate NK-cell-mediated tumor control. The IR T-cell immunoglobulin and ITIM domain (TIGIT) and its counter-receptor CD226 exert opposite effects on NK-cell-mediated tumor reactivity. EXPERIMENTAL DESIGN: We evaluated the frequency, phenotype, and functions of NK cells freshly isolated from healthy donors and patients with melanoma with multiparameter flow cytometry. We assessed TIGIT and CD226 cell surface expression and internalization upon binding to CD155. We evaluated the role of IL15 and TIGIT blockade in increasing NK-cell-mediated cytotoxicity in vitro and in two mouse models. RESULTS: NK cells are present at low frequencies in metastatic melanoma, are dysfunctional, and downregulate both TIGIT and CD226 expression. As compared with TIGIT- NK cells, TIGIT+ NK cells exhibit higher cytotoxic capacity and maturation, but paradoxically lower cytotoxicity against CD155+ MHC class I-deficient melanoma cells. Membrane bound CD155 triggers CD226 internalization and degradation, resulting in decreased NK-cell-mediated tumor reactivity. IL15 increases TIGIT and CD226 gene expression by tumor-infiltrating NK cells (TiNKs) and, together with TIGIT blockade, increases NK-cell-mediated melanoma cytotoxicity in vitro and decreases tumor metastasis in two mouse melanoma models. Specific deletion of TIGIT on transferred NK cells enhances the antimetastatic activity of IL15, while CD226 blockade decreases the effects of IL15 and TIGIT blockade. CONCLUSIONS: Our findings support the development of novel combinatorial immunotherapy with IL15 and TIGIT blockade to promote NK-cell-mediated destruction of MHC class I-deficient melanoma, which are refractory to CD8+ T-cell-mediated immunity.See related commentary by Pietra et al., p. 5274.


Asunto(s)
Interleucina-15/farmacología , Células Asesinas Naturales/inmunología , Melanoma/inmunología , Receptores Inmunológicos/genética , Animales , Antígenos de Diferenciación de Linfocitos T/genética , Antígenos de Diferenciación de Linfocitos T/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Interleucina-15/genética , Células Asesinas Naturales/efectos de los fármacos , Melanoma/sangre , Melanoma/genética , Melanoma/patología , Ratones , Receptores Inmunológicos/antagonistas & inhibidores , Receptores Virales/genética
10.
Cancer Immunol Res ; 7(8): 1258-1266, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31239318

RESUMEN

Multiple studies have associated the transcription factor IRF1 with tumor-suppressive activities. Here, we report an opposite tumor cell-intrinsic function of IRF1 in promoting tumor growth. IRF1-deficient tumor cells showed reduced tumor growth in MC38 and CT26 colon carcinoma and B16 melanoma mouse models. This reduction in tumor growth was dependent on host CD8+ T cells. Detailed profiling of tumor-infiltrating leukocytes did not show changes in the various T-cell and myeloid cell populations. However, CD8+ T cells that had infiltrated IRF1-deficieint tumors in vivo exhibited enhanced cytotoxicity. IRF1-deficient tumor cells lost the ability to upregulate PD-L1 expression in vitro and in vivo and were more susceptible to T-cell-mediated killing. Induced expression of PD-L1 in IRF1-deficient tumor cells restored tumor growth. These results indicate differential activity of IRF1 in tumor escape.


Asunto(s)
Antígeno B7-H1/genética , Regulación Neoplásica de la Expresión Génica , Inmunomodulación , Factor 1 Regulador del Interferón/metabolismo , Animales , Antígeno B7-H1/metabolismo , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Memoria Inmunológica , Inmunomodulación/genética , Factor 1 Regulador del Interferón/genética , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Melanoma Experimental , Ratones , Ratones Noqueados , Linfocitos T/inmunología , Linfocitos T/metabolismo
11.
JCI Insight ; 3(14)2018 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-30046006

RESUMEN

CD4+ Tregs impede T cell responses to tumors. They express multiple inhibitory receptors that support their suppressive functions, including T cell Ig and ITIM domain (TIGIT). In melanoma patients, we show that Tregs exhibit increased TIGIT expression and decreased expression of its competing costimulatory receptor CD226 as compared with CD4+ effector T cells, resulting in an increased TIGIT/CD226 ratio. Tregs failed to upregulate CD226 upon T cell activation. TIGIT+ Tregs are highly suppressive, stable, and enriched in tumors. TIGIT and CD226 oppose each other to augment or disrupt, respectively, Treg suppression and stability. A high TIGIT/CD226 ratio in Tregs correlates with increased Treg frequencies in tumors and poor clinical outcome upon immune checkpoint blockade. Altogether, our findings show that a high TIGIT/CD226 ratio in Tregs regulates their suppressive function and stability in melanoma. They provide the rationale for novel immunotherapies to activate CD226 in Tregs together with TIGIT blockade to counteract Treg suppression in cancer patients.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/metabolismo , Melanoma/metabolismo , Receptores Inmunológicos/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos , Citocinas , Humanos , Inmunofenotipificación , Inmunoterapia , Activación de Linfocitos , Melanoma/terapia , Linfocitos T Reguladores/inmunología , Proteínas Supresoras de Tumor/metabolismo
12.
J Clin Oncol ; : JCO1800632, 2018 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-30359157

RESUMEN

PURPOSE: Objective responses are reported in 34% to 37% of patients with programmed death-1 (PD-1)-naïve advanced melanoma treated with PD-1 inhibitors. Pre-existing CD8+ T-cell infiltrate and interferon (IFN) gene signature correlate with response to PD-1 blockade. Here, we report a phase Ib/II study of pembrolizumab/pegylated (PEG)-IFN combination in PD-1-naïve advanced melanoma. PATIENTS AND METHODS: PEG-IFN (1, 2, and 3 µg/kg per week) was dose escalated using a modified toxicity probability interval design in three cohorts of four patients each, whereas pembrolizumab was dosed at 2 mg/kg every 3 weeks in the phase Ib portion. Thirty-one patients were enrolled in the phase II portion. Primary objectives were safety and incidence of dose-limiting toxicities. Secondary objectives included objective response rate, progression-free survival (PFS), and overall survival. RESULTS: Forty-three patients with stage IV melanoma were enrolled in the phase Ib and II portions of the study and included in the analysis. At the data cutoff date (December 31, 2017), median follow-up duration was 25 months (range, 1 to 38 months). All 43 patients experienced at least one adverse event; grade 3/4 treatment-related adverse events occurred in 21 of 43 patients (48.8%). Objective responses were seen at all three dose levels among 43 evaluable patients. The objective response rate was 60.5%, with 46.5% of patients exhibiting ongoing response. Median PFS was 11.0 months in all patients and unreached in responders, whereas median overall survival remained unreached in all patients. The 2-year PFS rate was 46%. CONCLUSION: Pembrolizumab/PEG-IFN demonstrated an acceptable toxicity profile with promising evidence of clinical efficacy in PD-1-naïve metastatic melanoma. These results support the rationale to further investigate this pembrolizumab/PEG-IFN combination in this disease.

13.
Cancer Res ; 75(8): 1635-44, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25720800

RESUMEN

Immune checkpoint inhibitors show great promise as therapy for advanced melanoma, heightening the need to determine the most effective use of these agents. Here, we report that programmed death-1(high) (PD-1(high)) tumor antigen (TA)-specific CD8(+) T cells present at periphery and at tumor sites in patients with advanced melanoma upregulate IL10 receptor (IL10R) expression. Multiple subsets of peripheral blood mononucleocytes from melanoma patients produce IL10, which acts directly on IL10R(+) TA-specific CD8(+) T cells to limit their proliferation and survival. PD-1 blockade augments expression of IL10R by TA-specific CD8(+) T cells, thereby increasing their sensitivity to the immunosuppressive effects of endogenous IL10. Conversely, IL10 blockade strengthened the effects of PD-1 blockade in expanding TA-specific CD8(+) T cells and reinforcing their function. Collectively, our findings offer a rationale to block both IL10 and PD-1 to strengthen the counteraction of T-cell immunosuppression and to enhance the activity of TA-specific CD8(+) T cell in advanced melanoma patients.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Interleucina-10/fisiología , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma/inmunología , Receptor de Muerte Celular Programada 1/fisiología , Neoplasias Cutáneas/inmunología , Escape del Tumor , Anticuerpos/farmacología , Células Cultivadas , Humanos , Tolerancia Inmunológica , Interleucina-10/antagonistas & inhibidores , Interleucina-10/inmunología , Activación de Linfocitos , Melanoma/metabolismo , Melanoma/patología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Interleucina-10/metabolismo , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología
14.
J Clin Invest ; 125(5): 2046-58, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25866972

RESUMEN

T cell Ig and ITIM domain (TIGIT) is an inhibitory receptor expressed by activated T cells, Tregs, and NK cells. Here, we determined that TIGIT is upregulated on tumor antigen-specific (TA-specific) CD8⁺ T cells and CD8⁺ tumor-infiltrating lymphocytes (TILs) from patients with melanoma, and these TIGIT-expressing CD8⁺ T cells often coexpress the inhibitory receptor PD-1. Moreover, CD8⁺ TILs from patients exhibited downregulation of the costimulatory molecule CD226, which competes with TIGIT for the same ligand, supporting a TIGIT/CD226 imbalance in metastatic melanoma. TIGIT marked early T cell activation and was further upregulated by T cells upon PD-1 blockade and in dysfunctional PD-1⁺TIM-3⁺ TA-specific CD8⁺ T cells. PD-1⁺TIGIT⁺, PD-1⁻TIGIT⁺, and PD-1⁺TIGIT⁻ CD8⁺ TILs had similar functional capacities ex vivo, suggesting that TIGIT alone, or together with PD-1, is not indicative of T cell dysfunction. However, in the presence of TIGIT ligand-expressing cells, TIGIT and PD-1 blockade additively increased proliferation, cytokine production, and degranulation of both TA-specific CD8⁺ T cells and CD8⁺ TILs. Collectively, our results show that TIGIT and PD-1 regulate the expansion and function of TA-specific CD8⁺ T cells and CD8⁺ TILs in melanoma patients and suggest that dual TIGIT and PD-1 blockade should be further explored to elicit potent antitumor CD8⁺ T cell responses in patients with advanced melanoma.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma/inmunología , Proteínas de Neoplasias/fisiología , Receptor de Muerte Celular Programada 1/fisiología , Receptores Inmunológicos/fisiología , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Antígenos de Diferenciación de Linfocitos T/biosíntesis , Antígenos de Diferenciación de Linfocitos T/genética , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Citocinas/biosíntesis , Regulación Neoplásica de la Expresión Génica , Humanos , Memoria Inmunológica , Inmunofenotipificación , Subunidad beta del Receptor de Interleucina-2/biosíntesis , Subunidad beta del Receptor de Interleucina-2/genética , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor/metabolismo , Melanoma/patología , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/biosíntesis , Receptor de Muerte Celular Programada 1/genética , Receptores Inmunológicos/antagonistas & inhibidores , Receptores Inmunológicos/biosíntesis , Receptores Inmunológicos/genética , Receptores Virales/biosíntesis , Receptores Virales/genética , Especificidad del Receptor de Antígeno de Linfocitos T , Regulación hacia Arriba
15.
Cancer Res ; 74(4): 1045-55, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24343228

RESUMEN

Although melanoma vaccines stimulate tumor antigen-specific CD8(+) T cells, objective clinical responses are rarely observed. To investigate this discrepancy, we evaluated the character of vaccine-induced CD8(+) T cells with regard to the inhibitory T-cell coreceptors PD-1 and Tim-3 in patients with metastatic melanoma who were administered tumor vaccines. The vaccines included incomplete Freund's adjuvant, CpG oligodeoxynucleotide (CpG), and the HLA-A2-restricted analog peptide NY-ESO-1 157-165V, either by itself or in combination with the pan-DR epitope NY-ESO-1 119-143. Both vaccines stimulated rapid tumor antigen-specific CD8(+) T-cell responses detected ex vivo, however, tumor antigen-specific CD8(+) T cells produced more IFN-γ and exhibited higher lytic function upon immunization with MHC class I and class II epitopes. Notably, the vast majority of vaccine-induced CD8(+) T cells upregulated PD-1 and a minority also upregulated Tim-3. Levels of PD-1 and Tim-3 expression by vaccine-induced CD8(+) T cells at the time of vaccine administration correlated inversely with their expansion in vivo. Dual blockade of PD-1 and Tim-3 enhanced the expansion and cytokine production of vaccine-induced CD8(+) T cells in vitro. Collectively, our findings support the use of PD-1 and Tim-3 blockades with cancer vaccines to stimulate potent antitumor T-cell responses and increase the likelihood of clinical responses in patients with advanced melanoma.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Vacunas contra el Cáncer/uso terapéutico , Proliferación Celular , Activación de Linfocitos/genética , Melanoma/terapia , Proteínas de la Membrana/fisiología , Receptor de Muerte Celular Programada 1/fisiología , Linfocitos T CD4-Positivos/fisiología , Linfocitos T CD8-positivos/patología , Vacunas contra el Cáncer/inmunología , Adyuvante de Freund/uso terapéutico , Receptor 2 Celular del Virus de la Hepatitis A , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Humanos , Interferón gamma/metabolismo , Lípidos/uso terapéutico , Melanoma/genética , Melanoma/inmunología , Oligodesoxirribonucleótidos/uso terapéutico , Especificidad del Receptor de Antígeno de Linfocitos T , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA