Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Immunol Rev ; 302(1): 241-258, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34075584

RESUMEN

Cancer immunotherapies have rapidly changed the therapeutic landscape for cancer. Nevertheless, most of the patients show innate or acquired resistance to these therapies. Studies conducted in recent years have highlighted an emerging role of cancer-associated fibroblasts (CAFs) in immune regulation that shapes the tumor immune microenvironment (TIME) and influences response to cancer immunotherapies. In this review, we outline recent advances in the understanding of phenotypic and functional heterogeneity of CAFs. We will focus on emerging roles of CAFs in shaping the TIME, especially under a framework of tumor immunity continuum, and discuss current and future CAF-targeting therapeutic strategies in particular in the context of optimizing the success of immunotherapies.


Asunto(s)
Fibroblastos Asociados al Cáncer , Neoplasias , Humanos , Inmunoterapia , Neoplasias/terapia , Microambiente Tumoral
2.
Cancer Cell ; 39(7): 928-944.e6, 2021 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-33961783

RESUMEN

Distinct T cell infiltration patterns, i.e., immune infiltrated, excluded, and desert, result in different responses to cancer immunotherapies. However, the key determinants and biology underpinning these tumor immune phenotypes remain elusive. Here, we provide a high-resolution dissection of the entire tumor ecosystem through single-cell RNA-sequencing analysis of 15 ovarian tumors. Immune-desert tumors are characterized by unique tumor cell-intrinsic features, including metabolic pathways and low antigen presentation, and an enrichment of monocytes and immature macrophages. Immune-infiltrated and -excluded tumors differ markedly in their T cell composition and fibroblast subsets. Furthermore, our study reveals chemokine receptor-ligand interactions within and across compartments as potential mechanisms mediating immune cell infiltration, exemplified by the tumor cell-T cell cross talk via CXCL16-CXCR6 and stromal-immune cell cross talk via CXCL12/14-CXCR4. Our data highlight potential molecular mechanisms that shape the tumor immune phenotypes and may inform therapeutic strategies to improve clinical benefit from cancer immunotherapies.


Asunto(s)
Biomarcadores de Tumor/genética , Fibroblastos/inmunología , Neoplasias Ováricas/inmunología , Análisis de la Célula Individual/métodos , Células del Estroma/inmunología , Linfocitos T/inmunología , Microambiente Tumoral , Biomarcadores de Tumor/inmunología , Quimiocina CXCL12/genética , Quimiocina CXCL12/inmunología , Quimiocina CXCL16/genética , Quimiocina CXCL16/inmunología , Quimiocinas CXC/genética , Quimiocinas CXC/inmunología , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , RNA-Seq , Receptores CXCR4/genética , Receptores CXCR4/inmunología , Receptores CXCR6/genética , Receptores CXCR6/inmunología , Células del Estroma/metabolismo , Células del Estroma/patología , Linfocitos T/metabolismo , Linfocitos T/patología
3.
Nat Commun ; 11(1): 5583, 2020 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-33149148

RESUMEN

Close proximity between cytotoxic T lymphocytes and tumour cells is required for effective immunotherapy. However, what controls the spatial distribution of T cells in the tumour microenvironment is not well understood. Here we couple digital pathology and transcriptome analysis on a large ovarian tumour cohort and develop a machine learning approach to molecularly classify and characterize tumour-immune phenotypes. Our study identifies two important hallmarks characterizing T cell excluded tumours: 1) loss of antigen presentation on tumour cells and 2) upregulation of TGFß and activated stroma. Furthermore, we identify TGFß as an important mediator of T cell exclusion. TGFß reduces MHC-I expression in ovarian cancer cells in vitro. TGFß also activates fibroblasts and induces extracellular matrix production as a potential physical barrier to hinder T cell infiltration. Our findings indicate that targeting TGFß might be a promising strategy to overcome T cell exclusion and improve clinical benefits of cancer immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Carcinoma Epitelial de Ovario/inmunología , Regulación Neoplásica de la Expresión Génica/inmunología , Neoplasias Ováricas/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral/inmunología , Presentación de Antígeno/inmunología , Carcinoma Epitelial de Ovario/genética , Carcinoma Epitelial de Ovario/metabolismo , Carcinoma Epitelial de Ovario/patología , Línea Celular Tumoral , Estudios de Cohortes , Metilación de ADN , Endopeptidasas , Femenino , Gelatinasas/metabolismo , Perfilación de la Expresión Génica , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Aprendizaje Automático , Proteínas de la Membrana/metabolismo , Familia de Multigenes , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Pronóstico , RNA-Seq , Serina Endopeptidasas/metabolismo , Células del Estroma/metabolismo
4.
J Immunother Cancer ; 8(2)2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32784216

RESUMEN

BACKGROUND: Oxazaphosphorines (cyclophosphamide (CPA), ifosfamide (IFO)) are major alkylating agents of polychemotherapy protocols but limiting their toxicity and increasing their efficacy could be of major interest. Oxazaphosphorines are prodrugs that require an activation by cytochrome P450 (CYP). CPA is mainly metabolized (>80%) to phosphoramide mustard while only 10%-50% of IFO is transformed in the alkylating entity, isophosphoramide mustard and 50%-90% of IFO release chloroacetaldehyde, a nephrotoxic and neurotoxic metabolite. Geranyloxy-IFO (G-IFO) was reported as a preactivated IFO to circumvent the toxic pathway giving directly the isophosphoramide mustard without CYP metabolization. The similarity in structure of CPA and IFO and the similarity in metabolic balance of CPA and G-IFO have led us to explore immunomodulatory effect of these components in mice and to investigate the combination of these oxazaphosphorines with immune checkpoint blockers (ICB). METHODS: The investigation of the immunomodulatory properties of IFO and G-IFO compared with CPA has been conducted through immune cell phenotyping by flow cytometry and analysis of the cytokine profile of T cells after ex-vivo restimulation. T cell-mediated antitumor efficacy was confirmed in CD4+ and CD8+ T cell-depleted mice. A combination of oxazaphosphorines with an anti-programmed cell death 1 (PD-1) antibody has been studied in MCA205 tumor-bearing mice. RESULTS: Studies on a MCA205 mouse model have demonstrated a dose-dependent effect of IFO and G-IFO on T cell immunity. These components in particular favored Th1 polarization when used at low dose (150 and eq. 100 mg/kg, respectively). Antitumor activity at low dose was abolished in mice depleted in CD4+ and CD8+ T cells. G-IFO at low dose (eq. 100 mg/kg) in combination with anti-PD-1 antidody showed high synergistic antitumor efficacy compared with IFO. CONCLUSION: Oxazaphosphorines are characterized by a dual mechanism of antitumor action; low-dose schedules should be preferred in combination with ICB, and dose escalation was found to have better utility in polychemotherapy protocols where a conventional direct cytotoxic anticancer effect is needed. G-IFO, the novel oxazaphosphorine drug, has shown a better metabolic index compared with IFO as its metabolization gives mainly the alkylating mustard as CPA (and not IFO) and a best potential in combination with ICB.


Asunto(s)
Antineoplásicos/uso terapéutico , Ciclofosfamida/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Mostazas de Fosforamida/uso terapéutico , Animales , Antineoplásicos/farmacología , Ciclofosfamida/farmacología , Modelos Animales de Enfermedad , Femenino , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Ratones , Mostazas de Fosforamida/farmacología
5.
J Immunother Cancer ; 8(1)2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32532840

RESUMEN

BACKGROUND: As the immune system is compromised in patients with cancer, therapeutic strategies to stimulate immunity appear promising, to avoid relapse and increase long-term overall survival. Interleukin-15 (IL-15) has similar properties to IL-2, but does not cause activation-induced cell death nor activation and proliferation of regulatory T cells (Treg), which makes it a serious candidate for anticancer immunotherapy. However, IL-15 has a short half-life and high doses are needed to achieve responses. Designed to enhance its activity, receptor-linker-IL-15 (RLI) (SO-C101) is a fusion molecule of human IL-15 covalently linked to the human IL-15Rα sushi+ domain currently assessed in a phase I/Ib clinical trial on patients with advanced/metastatic solid cancer. METHODS: We investigated the antimetastatic activity of RLI in a 4T1 mouse mammary carcinoma that spontaneously metastasizes and evaluated its immunomodulatory role in the metastatic lung microenvironment. We further characterized the proliferation, maturation and cytotoxic functions of natural killer (NK) cells in tumor-free mice treated with RLI. Finally, we explored the effect of RLI on human NK cells from healthy donors and patients with non-small cell lung cancer (NSCLC). RESULTS: RLI treatment displayed antimetastatic properties in the 4T1 mouse model. By characterizing the lung microenvironment, we observed that RLI restored the balance between NK cells and neutrophils (CD11b+ Ly6Ghigh Ly6Clow) that massively infiltrate lungs of 4T1-tumor bearing mice. In addition, the ratio between NK cells and Treg was strongly increased by RLI treatment. Further pharmacodynamic studies in tumor-free mice revealed superior proliferative and cytotoxic functions on NK cells after RLI treatment compared with IL-15 alone. Characterization of the maturation stage of NK cells demonstrated that RLI favored accumulation of CD11b+ CD27high KLRG1+ mature NK cells. Finally, RLI demonstrated potent immunostimulatory properties on human NK cells by inducing proliferation and activation of NK cells from healthy donors and enhancing cytotoxic responses to NKp30 crosslinking in NK cells from patients with NSCLC. CONCLUSIONS: Collectively, our work demonstrates superior activity of RLI compared with rhIL-15 in modulating and activating NK cells and provides additional evidences for a therapeutic strategy using RLI as antimetastatic molecule.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Interleucina-15/administración & dosificación , Células Asesinas Naturales/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Proteínas Recombinantes de Fusión/administración & dosificación , Animales , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Línea Celular Tumoral/trasplante , Femenino , Voluntarios Sanos , Humanos , Interleucina-15/agonistas , Células Asesinas Naturales/inmunología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Ratones , Cultivo Primario de Células , Proteínas Recombinantes/administración & dosificación , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
6.
Nat Commun ; 11(1): 2168, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32358520

RESUMEN

Gut microbiota composition influences the clinical benefit of immune checkpoints in patients with advanced cancer but mechanisms underlying this relationship remain unclear. Molecular mechanism whereby gut microbiota influences immune responses is mainly assigned to gut microbial metabolites. Short-chain fatty acids (SCFA) are produced in large amounts in the colon through bacterial fermentation of dietary fiber. We evaluate in mice and in patients treated with anti-CTLA-4 blocking mAbs whether SCFA levels is related to clinical outcome. High blood butyrate and propionate levels are associated with resistance to CTLA-4 blockade and higher proportion of Treg cells. In mice, butyrate restrains anti-CTLA-4-induced up-regulation of CD80/CD86 on dendritic cells and ICOS on T cells, accumulation of tumor-specific T cells and memory T cells. In patients, high blood butyrate levels moderate ipilimumab-induced accumulation of memory and ICOS + CD4 + T cells and IL-2 impregnation. Altogether, these results suggest that SCFA limits anti-CTLA-4 activity.


Asunto(s)
Biomarcadores de Tumor/sangre , Antígeno CTLA-4/antagonistas & inhibidores , Antígeno CTLA-4/metabolismo , Ácidos Grasos Volátiles/sangre , Neoplasias/sangre , Neoplasias/metabolismo , Animales , Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Biomarcadores de Tumor/metabolismo , Butiratos/sangre , Células Dendríticas/metabolismo , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/genética , Humanos , Ipilimumab/farmacología , Ratones , Ratones Endogámicos BALB C , Propionatos/sangre , ARN Ribosómico 16S/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Linfocitos T Reguladores/metabolismo
7.
J Immunol ; 197(1): 168-78, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27217584

RESUMEN

Tumors with the help of the surrounding environment facilitate the immune suppression in patients, and immunotherapy can counteract this inhibition. Among immunotherapeutic strategies, the immunostimulatory cytokine IL-15 could represent a serious candidate for the reactivation of antitumor immunity. However, exogenous IL-15 may have a limited impact on patients with cancer due to its dependency on IL-15Rα frequently downregulated in cancer patients. In this work, we studied the antitumor activity of the IL-15 superagonist receptor-linker-IL-15 (RLI), designed to bypass the need of endogenous IL-15Rα. RLI consists of human IL-15 covalently linked to the human IL-15Rα sushi(+) domain. In a mouse model of colorectal carcinoma, RLI as a stand-alone treatment could limit tumor outgrowth only when initiated at an early time of tumor development. At a later time, RLI was not effective, coinciding with the strong accumulation of terminally exhausted programmed cell death-1 (PD-1)(high) T cell Ig mucin-3(+) CD8(+) T cells, suggesting that RLI was not able to reactivate terminally exhausted CD8(+) T cells. Combination with PD-1 blocking Ab showed synergistic activity with RLI, but not with IL-15. RLI could induce a greater accumulation of memory CD8(+) T cells and a stronger effector function in comparison with IL-15. Ex vivo stimulation of tumor-infiltrated lymphocytes from 16 patients with renal cell carcinoma demonstrated 56% of a strong tumor-infiltrated lymphocyte reactivation with the combination anti-PD-1/RLI compared with 43 and 6% with RLI or anti-PD-1, respectively. Altogether, this work provides evidence that the sushi-IL-15Rα/IL-15 fusion protein RLI enhances antitumor activity of anti-PD-1 treatment and is a promising approach to stimulate host immunity.


Asunto(s)
Adenocarcinoma/terapia , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Linfocitos T CD8-positivos/efectos de los fármacos , Carcinoma de Células Renales/terapia , Neoplasias del Colon/terapia , Inmunoterapia/métodos , Interleucina-15/metabolismo , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/uso terapéutico , Adenocarcinoma/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Carcinoma de Células Renales/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/inmunología , Citotoxicidad Inmunológica/efectos de los fármacos , Sinergismo Farmacológico , Quimioterapia Combinada , Humanos , Memoria Inmunológica/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Factor de Transcripción STAT5/metabolismo , Transducción de Señal , Carga Tumoral/efectos de los fármacos
9.
Bull Cancer ; 102(1): 34-52, 2015 Jan.
Artículo en Francés | MEDLINE | ID: mdl-25609492

RESUMEN

The discovery and understanding of complex cellular interactions that govern the immune system contributed to the pharmacological targeting of anti-tumor immunity. The activity of immune effector cells, such as NK and T-cells, is regulated by a wide range of activating and inhibiting receptors or ligands. Drugs that target these receptors or ligands can modulate the immune response by exerting antagonistic or agonistic effects. Over the past decade, several immunomodulators have demonstrated clinical effectiveness, and three of them have already been approved for use in oncology. The development of these immunotherapy approaches presented unique challenges for safety and efficacy, requiring revising clinical response criteria and the establishment of guidelines to help oncologists to manage properly inflammatory toxicities. The introduction of these immunotherapies is a revolution in oncology. However, additional efforts in terms of optimizing treatment administration and identification of biomarkers are needed. Identifying the immunodynamics of various immunomodulators should allow a better understanding of anti-tumor and inflammatory mechanisms, and certainly give the opportunity to develop effective therapeutic combinations without potentiating adverse events.


Asunto(s)
Tolerancia Inmunológica , Factores Inmunológicos/uso terapéutico , Inmunoterapia , Neoplasias/inmunología , Neoplasias/terapia , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antígenos de Neoplasias/inmunología , Antígeno CTLA-4/inmunología , Humanos , Indoles/uso terapéutico , Ipilimumab , Células Asesinas Naturales/fisiología , Melanoma/inmunología , Melanoma/terapia , Nivolumab , Sulfonamidas/uso terapéutico , Linfocitos T/fisiología , Microambiente Tumoral/inmunología , Vemurafenib
10.
Cell Res ; 25(2): 208-24, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25582080

RESUMEN

The cytotoxic T lymphocyte antigen-4 (CTLA-4)-blocking antibody ipilimumab induces immune-mediated long-term control of metastatic melanoma in a fraction of patients. Although ipilimumab undoubtedly exerts its therapeutic effects via immunostimulation, thus far clinically useful, immunologically relevant biomarkers that predict treatment efficiency have been elusive. Here, we show that neutralization of IL-2 or blocking the α and ß subunits of the IL-2 receptor (CD25 and CD122, respectively) abolished the antitumor effects and the accompanying improvement of the ratio of intratumoral T effector versus regulatory cells (Tregs), which were otherwise induced by CTLA-4 blockade in preclinical mouse models. CTLA-4 blockade led to the reduction of a suppressive CD4(+) T cell subset expressing Lag3, ICOS, IL-10 and Egr2 with a concomitant rise in IL-2-producing effector cells that lost FoxP3 expression and accumulated in regressing tumors. While recombinant IL-2 improved the therapeutic efficacy of CTLA-4 blockade, the decoy IL-2 receptor α (IL-2Rα, sCD25) inhibited the anticancer effects of CTLA-4 blockade. In 262 metastatic melanoma patients receiving ipilimumab, baseline serum concentrations of sCD25 represented an independent indicator of overall survival, with high levels predicting resistance to therapy. Altogether, these results unravel a role for IL-2 and IL-2 receptors in the anticancer activity of CTLA-4 blockade. Importantly, our study provides the first immunologically relevant biomarker, namely elevated serum sCD25, that predicts resistance to CTLA-4 blockade in patients with melanoma.


Asunto(s)
Antígeno CTLA-4/metabolismo , Subunidad alfa del Receptor de Interleucina-2/sangre , Melanoma/patología , Receptores de Interleucina-2/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Antígeno CTLA-4/inmunología , Estudios de Cohortes , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunoterapia , Subunidad alfa del Receptor de Interleucina-2/inmunología , Ipilimumab , Masculino , Melanoma/mortalidad , Melanoma/terapia , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Receptores de Interleucina-2/genética , Receptores de Interleucina-2/inmunología , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Linfocitos T Reguladores/inmunología , Regulación hacia Arriba/efectos de los fármacos , Adulto Joven
11.
Cancer Res ; 73(12): 3499-510, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23592754

RESUMEN

Cancer immunosurveillance relies on effector/memory tumor-infiltrating CD8(+) T cells with a T-helper cell 1 (TH1) profile. Evidence for a natural killer (NK) cell-based control of human malignancies is still largely missing. The KIT tyrosine kinase inhibitor imatinib mesylate markedly prolongs the survival of patients with gastrointestinal stromal tumors (GIST) by direct effects on tumor cells as well as by indirect immunostimulatory effects on T and NK cells. Here, we investigated the prognostic value of tumor-infiltrating lymphocytes (TIL) expressing CD3, Foxp3, or NKp46 (NCR1) in a cohort of patients with localized GIST. We found that CD3(+) TIL were highly activated in GIST and were especially enriched in areas of the tumor that conserve class I MHC expression despite imatinib mesylate treatment. High densities of CD3(+) TIL predicted progression-free survival (PFS) in multivariate analyses. Moreover, GIST were infiltrated by a homogeneous subset of cytokine-secreting CD56(bright) (NCAM1) NK cells that accumulated in tumor foci after imatinib mesylate treatment. The density of the NK infiltrate independently predicted PFS and added prognostic information to the Miettinen score, as well as to the KIT mutational status. NK and T lymphocytes preferentially distributed to distinct areas of tumor sections and probably contributed independently to GIST immunosurveillance. These findings encourage the prospective validation of immune biomarkers for optimal risk stratification of patients with GIST.


Asunto(s)
Tumores del Estroma Gastrointestinal/inmunología , Células Asesinas Naturales/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos T/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Benzamidas/inmunología , Benzamidas/uso terapéutico , Complejo CD3/inmunología , Complejo CD3/metabolismo , Antígeno CD56/inmunología , Antígeno CD56/metabolismo , Estudios de Cohortes , Femenino , Citometría de Flujo , Factores de Transcripción Forkhead/inmunología , Factores de Transcripción Forkhead/metabolismo , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Tumores del Estroma Gastrointestinal/metabolismo , Humanos , Mesilato de Imatinib , Inmunohistoquímica , Estimación de Kaplan-Meier , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/metabolismo , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/metabolismo , Masculino , Persona de Mediana Edad , Análisis Multivariante , Receptor 1 Gatillante de la Citotoxidad Natural/inmunología , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Piperazinas/inmunología , Piperazinas/uso terapéutico , Pronóstico , Inhibidores de Proteínas Quinasas/inmunología , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/inmunología , Pirimidinas/uso terapéutico , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Resultado del Tratamiento
12.
Oncoimmunology ; 2(2): e23080, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23525357

RESUMEN

We performed a Phase I clinical trial from October 2007 to October 2009, enrolling patients affected by refractory solid tumors, to determine the maximum tolerated dose (MTD) of interleukin (IL)-2 combined with low dose cyclophosphamide (CTX) and imatinib mesylate (IM). In a companion paper published in this issue of OncoImmunology, we show that the MTD of IL-2 is 6 MIU/day for 5 consecutive days, and that IL-2 increases the impregnation of both IM and of its main metabolite, CGP74588. Among the secondary objectives, we wanted to determine immunological markers that might be associated with progression-free survival (PFS) and/or overall survival (OS). The combination therapy markedly reduced the absolute counts of B, CD4+ T and CD8+ T cells in a manner that was proportional to IL-2 dose. There was a slight (less than 2-fold) increase in the proportion of regulatory T cells (Tregs) among CD4+ T cells in response to IM plus IL-2. The natural killer (NK)-cell compartment was activated, exhibiting a significant upregulation of HLA-DR, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and CD56. The abundance of HLA-DR+ NK cells after one course of combination therapy positively correlated with both PFS and OS. The IL-2-induced rise of the CD4+:CD8+ T-cell ratio calculated after the first cycle of treatment was also positively associated with OS. Overall, the combination of IM and IL-2 promoted the rapid expansion of HLA-DR+ NK cells and increased the CD4+:CD8+ T-cell ratio, both being associated with clinical benefits. This combinatorial regimen warrants further investigation in Phase II clinical trials, possibly in patients affected by gastrointestinal stromal tumors, a setting in which T and NK cells may play an important therapeutic role.

13.
Front Immunol ; 3: 395, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23269924

RESUMEN

Natural killer (NK) cells belong to the innate immune system and were initially described functionallywise by their spontaneous cytotoxic potential against transformed or virus-infected cells. A delicate balance between activating and inhibiting receptors regulates NK cell tolerance. A better understanding of tissue resident NK cells, of NK cell maturation stages and migration patterns has evolved allowing a thoughtful evaluation of their modus operandi. While evidence has been brought up for their relevance as gate keepers in some hematopoietic malignancies, the role of NK cells against progression and dissemination of solid tumors remains questionable. Hence, many studies pointed out the functional defects of the rare NK cell infiltrates found in tumor beds and the lack of efficacy of adoptively transferred NK cells in patients. However, several preclinical evidences suggest their anti-metastatic role in a variety of mouse tumor models. In the present review, we discuss NK cell functions according to their maturation stage and environmental milieu, the receptor/ligand interactions dictating tumor cell recognition and recapitulate translational studies aimed at deciphering their prognostic or predictive role against human solid malignancies.

14.
Cancer Res ; 72(11): 2757-67, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22427351

RESUMEN

During cancer development, a number of regulatory cell subsets and immunosuppressive cytokines subvert adaptive immune responses. Although it has been shown that tumor-derived interleukin (IL)-18 participates in the PD-1-dependent tumor progression in NK cell-controlled cancers, the mechanistic cues underlying this immunosuppression remain unknown. Here, we show that IL-18 converts a subset of Kit(-) (CD11b(-)) into Kit(+) natural killer (NK) cells, which accumulate in all lymphoid organs of tumor bearers and mediate immunoablative functions. Kit(+) NK cells overexpressed B7-H1/PD-L1, a ligand for PD-1. The adoptive transfer of Kit(+) NK cells promoted tumor growth in two pulmonary metastases tumor models and significantly reduced the dendritic and NK cell pools residing in lymphoid organs in a B7-H1-dependent manner. Neutralization of IL-18 by RNA interference in tumors or systemically by IL-18-binding protein dramatically reduced the accumulation of Kit(+)CD11b(-) NK cells in tumor bearers. Together, our findings show that IL-18 produced by tumor cells elicits Kit(+)CD11b(-) NK cells endowed with B7-H1-dependent immunoablative functions in mice.


Asunto(s)
Interleucina-18/inmunología , Células Asesinas Naturales/inmunología , Neoplasias/inmunología , Animales , Antígeno B7-H1/análisis , Antígeno B7-H1/fisiología , Antígeno CD11b/análisis , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-kit/análisis
15.
Cancer Res ; 71(16): 5393-9, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21724589

RESUMEN

Immunosuppressive cytokines subvert innate and adaptive immune responses during cancer progression. The inflammatory cytokine interleukin-18 (IL-18) is known to accumulate in cancer patients, but its pathophysiological role remains unclear. In this study, we show that low levels of circulating IL-18, either exogenous or tumor derived, act to suppress the NK cell arm of tumor immunosurveillance. IL-18 produced by tumor cells promotes the development of NK-controlled metastases in a PD-1-dependent manner. Accordingly, PD-1 is expressed by activated mature NK cells in lymphoid organs of tumor bearers and is upregulated by IL-18. RNAi-mediated knockdown of IL-18 in tumors, or its systemic depletion by IL-18-binding protein, are sufficient to stimulate NK cell-dependent immunosurveillance in various tumor models. Together, these results define IL-18 as an immunosuppressive cytokine in cancer. Our findings suggest novel clinical implementations of anti-PD-1 antibodies in human malignancies that produce IL-18.


Asunto(s)
Antígenos de Superficie/fisiología , Proteínas Reguladoras de la Apoptosis/fisiología , Interleucina-18/fisiología , Melanoma Experimental/inmunología , Animales , Antígenos de Superficie/inmunología , Proteínas Reguladoras de la Apoptosis/inmunología , Autoanticuerpos/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Tolerancia Inmunológica , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Metástasis de la Neoplasia/inmunología , Receptor de Muerte Celular Programada 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...