Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Oncoimmunology ; 10(1): 1935557, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34239773

RESUMEN

Cancer immunotherapies have induced long-lasting responses in cancer patients including those with melanoma and head and neck squamous cell carcinoma (HNSCC). However, the majority of treated patients does not achieve clinical benefit from immunotherapy because of systemic tumor-induced immunosuppression. Monocytic myeloid-derived suppressor cells (M-MDSCs) are implicated as key players in inhibiting anti-tumor immune responses and their frequencies are closely associated with tumor progression. Tumor-derived signals, including signaling via STAT3-COX-2, induce the transformation of monocytic precursors into suppressive M-MDSCs. In a retrospective assessment, we observed that survival of melanoma patients undergoing dendritic cell vaccination was negatively associated with blood M-MDSC levels. Previously, it was shown that platinum-based chemotherapeutics inhibit STAT signaling. Here, we show that cisplatin and oxaliplatin treatment interfere with the development of M-MDSCs, potentially synergizing with cancer immunotherapy. In vitro, subclinical doses of platinum-based drugs prevented the generation of COX-2+ M-MDSCs induced by tumor cells from melanoma patients. This was confirmed in HNSCC patients where intravenous cisplatin treatment drastically lowered M-MDSC frequency while monocyte levels remained stable. In treated patients, expression of COX-2 and arginase-1 in M-MDSCs was significantly decreased after two rounds of cisplatin, indicating inhibition of STAT3 signaling. In line, the capacity of M-MDSCs to inhibit activated T cell responses ex vivo was significantly decreased after patients received cisplatin. These results show that platinum-based chemotherapeutics inhibit the expansion and suppressive activity of M-MDSCs in vitro and in cancer patients. Therefore, platinum-based drugs have the potential to enhance response rates of immunotherapy by overcoming M-MDSC-mediated immunosuppression.


Asunto(s)
Melanoma , Células Supresoras de Origen Mieloide , Cisplatino/farmacología , Humanos , Melanoma/tratamiento farmacológico , Monocitos , Estudios Retrospectivos
2.
Cancer Immunol Immunother ; 69(3): 477-488, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31980913

RESUMEN

BACKGROUND: Autologous dendritic cell (DC) vaccines can induce tumor-specific T cells, but their effect can be counteracted by immunosuppressive mechanisms. Cisplatin has shown immunomodulatory effects in vivo which may enhance efficacy of DC vaccination. METHODS: This is a prospective, randomized, open-label phase 2 study (NCT02285413) including stage III and IV melanoma patients receiving 3 biweekly vaccinations of gp100 and tyrosinase mRNA-loaded monocyte-derived DCs with or without cisplatin. Primary objectives were to study immunogenicity and feasibility, and secondary objectives were to assess toxicity and survival. RESULTS: Twenty-two stage III and 32 stage IV melanoma patients were analyzed. Antigen-specific CD8+ T cells were found in 44% versus 67% and functional T cell responses in 28% versus 19% of skin-test infiltrating lymphocytes in patients receiving DC vaccination with and without cisplatin, respectively. Four patients stopped cisplatin because of toxicity and continued DC monotherapy. No therapy-related grade 3 or 4 adverse events occurred due to DC monotherapy. During combination therapy, one therapy-related grade 3 adverse event, decompensated heart failure due to fluid overload, occurred. The clinical outcome parameters did not clearly suggest significant differences. CONCLUSIONS: Combination of DC vaccination and cisplatin in melanoma patients is feasible and safe, but does not seem to result in more tumor-specific T cell responses or improved clinical outcome, when compared to DC vaccination monotherapy.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Cisplatino/uso terapéutico , Células Dendríticas/inmunología , Melanoma/tratamiento farmacológico , Adolescente , Adulto , Anciano , Vacunas contra el Cáncer/farmacología , Cisplatino/farmacología , Femenino , Humanos , Masculino , Melanoma/patología , Persona de Mediana Edad , Monocitos/inmunología , Estadificación de Neoplasias , Estudios Prospectivos , Vacunación , Adulto Joven
3.
J Immunol Res ; 2019: 7458238, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31309123

RESUMEN

The STAT signaling pathway is important in dendritic cell (DC) development and function. Tumor cells can induce STAT signaling, thereby inhibiting DC maturation and immunostimulatory functions, leading to hampered efficacy of DC-based immunotherapies. Platinum-based chemotherapeutics can inhibit STAT signaling, thereby making them an interesting tool to improve DC development and function. In this study, we provide a comprehensive overview of STAT expression and phosphorylation during DC differentiation and maturation and investigate the effects of platinum drugs on STAT signaling during these processes. Monocytes were differentiated into monocyte-derived DCs (moDCs) with IL-4 and GM-CSF and matured with cytokines or TLR ligands. STAT expression and phosphorylation were analyzed by western blotting, and moDC viability and phenotype were analyzed by flow cytometry. Platinum drugs were added at day 3 of differentiation or at the start of maturation to investigate regulation of the STAT signaling pathway. All STAT proteins were expressed during moDC differentiation and STAT1, STAT5, and STAT6 were phosphorylated. No significant changes occurred in the expression and phosphorylation state of the STAT proteins during differentiation. After maturation with TLR ligands, the expression of STAT1 increased, but other STAT proteins were not affected. Phosphorylation of STAT1 and STAT3 increased during maturation, where TLR ligands induced significantly higher levels of phosphorylation than cytokines. Platinum drugs cisplatin and oxaliplatin significantly inhibited phosphorylation of STAT6 during differentiation and maturation. Treatment did not affect the phenotype or viability of the cells. As STAT6 is an important regulator of DC function, these findings suggest a role for platinum-based chemotherapeutics to enhance DC function via inhibition of STAT signaling, thereby potentially enhancing efficacy of DC-based immunotherapies.


Asunto(s)
Antineoplásicos/farmacología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Platino (Metal)/farmacología , Factores de Transcripción STAT/metabolismo , Biomarcadores , Diferenciación Celular , Células Dendríticas/citología , Células Dendríticas/inmunología , Expresión Génica , Humanos , Inmunofenotipificación , Fosforilación , Factores de Transcripción STAT/genética , Transducción de Señal
4.
Oncotarget ; 8(40): 67439-67456, 2017 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-28978044

RESUMEN

Immunotherapy for metastatic melanoma offers great promise but, to date, only a subset of patients have responded. There is an urgent need to identify ways of allocating patients to the most beneficial therapy, to increase survival and decrease therapy-associated morbidity and costs. Blood-based biomarkers are of particular interest because of their straightforward implementation in routine clinical care. We sought to identify markers for dendritic cell (DC) vaccine-based immunotherapy against metastatic melanoma through gene expression analysis of peripheral blood mononuclear cells. A large-scale microarray analysis of 74 samples from two treatment centers, taken directly after the first round of DC vaccination, was performed. We found that phosphatidylethanolamine binding protein 1 (PEBP1)/Raf Kinase inhibitory protein (RKIP) expression can be used to identify a significant proportion of patients who performed poorly after DC vaccination. This result was validated by q-PCR analysis on blood samples from a second cohort of 95 patients treated with DC vaccination in four different centers. We conclude that low PEBP1 expression correlates with poor overall survival after DC vaccination. Intriguingly, this was only the case for expression of PEBP1 after, but not prior to, DC vaccination. Moreover, the change in PEBP1 expression upon vaccination correlated well with survival. Further analyses revealed that PEBP1 expression positively correlated with genes involved in T cell responses but inversely correlated with genes associated with myeloid cells and aberrant inflammation including STAT3, NOTCH1, and MAPK1. Concordantly, PEBP1 inversely correlated with the myeloid/lymphoid-ratio and was suppressed in patients suffering from chronic inflammatory disease.

5.
Oncotarget ; 8(33): 54434-54443, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28903353

RESUMEN

Platinum-based chemotherapeutics are amongst the most powerful anti-cancer drugs. Although their exact mechanism of action is not well understood, it is thought to be mediated through covalent DNA binding. We investigated the effect of platinum-based chemotherapeutics on signaling through signal transducer and activator of transcription (STAT) proteins, which are involved in many oncogenic signaling pathways. We performed in vitro experiments in various cancer cell lines, investigating the effects of platinum chemotherapeutics on STAT phosphorylation and nuclear translocation, the expression of STAT-modulating proteins and downstream signaling pathways. Direct binding of platinum to STAT proteins was assessed using an AlphaScreen assay. Nuclear STAT3 expression was determined by immunohistochemistry and correlated with disease-free survival in retrospective cohorts of head and neck squamous cell carcinoma (HNSCC) patients treated with cisplatin-based chemoradiotherapy (n= 65) or with radiotherapy alone (n = 32). At clinically relevant concentrations, platinum compounds inhibited STAT phosphorylation, resulting in loss of constitutively activated STAT proteins in multiple distinct cancer cell lines. Platinum drugs specifically inhibited phospho-tyrosine binding to SH2 domains, thereby blocking STAT activation, and subsequently downregulating pro-survival- and anti-apoptotic- target genes. Importantly, we found that active STAT3 in tumors directly correlated with response to cisplatin-based chemoradiotherapy in HNSCC patients (p = 0.006). These findings provide insight into a novel, non-DNA-targeted mechanism of action of platinum drugs, and could be leveraged into the use of STAT expression as predictive biomarker for cisplatin chemotherapy and to potentiate other therapeutic strategies such as immunotherapy.

6.
Oncoimmunology ; 5(7): e1196312, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27622051

RESUMEN

Cancer immunotherapy is a promising therapeutic avenue; however, in practice its efficacy is hampered by an immunosuppressive tumor microenvironment that consists of suppressive cell types like myeloid-derived suppressor cells (MDSCs). Eradication or reprogramming of MDSCs could therefore enhance clinical responses to immunotherapy. Here, we review clinically available drugs that target MDSCs, often through inhibition of STAT signaling, which is essential for MDSC accumulation and suppressive functions. Interestingly, several drugs used for non-cancerous indications and natural compounds similarly inhibit MDSCs by STAT inhibition, but have fewer side effects than anticancer drugs. Therefore, they show great potential for combination strategies with immunotherapy.

7.
Oncoimmunology ; 5(8): e1192739, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27622063

RESUMEN

Chemotherapeutics, including the platinum compounds oxaliplatin (OXP) and cisplatin (CDDP), are standard care of treatment for cancer. Although chemotherapy has long been considered immunosuppressive, evidence now suggests that certain cytotoxic agents can efficiently stimulate antitumor responses, through the induction of a form of apoptosis, called immunogenic cell death (ICD). ICD is characterized by exposure of calreticulin and heat shock proteins (HSPs), secretion of ATP and release of high-mobility group box 1 (HMGB1). Proper activation of the immune system relies on the integration of these signals by dendritic cells (DCs). Studies on the crucial role of DCs, in the context of ICD, have been performed using mouse models or human in vitro-generated monocyte-derived DCs (moDCs), which do not fully recapitulate the in vivo situation. Here, we explore the effect of platinum-induced ICD on phenotype and function of human blood circulating DCs. Tumor cells were treated with OXP or CDDP and induction of ICD was investigated. We show that both platinum drugs triggered translocation of calreticulin and HSP70, as well as the release of ATP and HMGB1. Platinum treatment increased phagocytosis of tumor fragments by human blood DCs and enhanced phenotypic maturation of blood myeloid and plasmacytoid DCs. Moreover, upon interaction with platinum-treated tumor cells, CD1c(+) DCs efficiently stimulated allogeneic proliferation of T lymphocytes. Together, our observations indicate that platinum-treated tumor cells may exert an active stimulatory effect on human blood DCs. In particular, these data suggest that CD1c(+) DCs are critical mediators of immune responses induced by ICD.

8.
Cancer Res ; 76(15): 4332-46, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27325645

RESUMEN

The tumor microenvironment is characterized by regulatory T cells, type II macrophages, myeloid-derived suppressor cells, and other immunosuppressive cells that promote malignant progression. Here we report the identification of a novel BDCA1(+)CD14(+) population of immunosuppressive myeloid cells that are expanded in melanoma patients and are present in dendritic cell-based vaccines, where they suppress CD4(+) T cells in an antigen-specific manner. Mechanistic investigations showed that BDCA1(+)CD14(+) cells expressed high levels of the immune checkpoint molecule PD-L1 to hinder T-cell proliferation. While this BDCA1(+)CD14(+) cell population expressed markers of both BDCA1(+) dendritic cells and monocytes, analyses of function, transcriptome, and proteome established their unique nature as exploited by tumors for immune escape. We propose that targeting these cells may improve the efficacy of cancer immunotherapy. Cancer Res; 76(15); 4332-46. ©2016 AACR.


Asunto(s)
Vacunas contra el Cáncer/genética , Células Dendríticas/inmunología , Receptores de Lipopolisacáridos/metabolismo , Melanoma/genética , Células Progenitoras Mieloides/metabolismo , Proliferación Celular , Humanos , Melanoma/patología , Microambiente Tumoral
9.
J Transl Med ; 14: 88, 2016 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-27075584

RESUMEN

BACKGROUND: Melanoma is the most lethal type of skin cancer and its incidence is progressively increasing. The introductions of immunotherapy and targeted therapies have tremendously improved the treatment of melanoma. Selective inhibition of BRAF by vemurafenib results in objective clinical responses in around 50 % of patients suffering from BRAFV600 mutated melanoma. However, drug resistance often results in hampering long-term tumor control. Alternatively, immunotherapy by vaccination with natural dendritic cells (nDCs) demonstrated long-term tumor control in a proportion of patients. We postulate that the rapid tumor debulking by vemurafenib can synergize the long-term tumor control of nDC vaccination to result in an effective treatment modality in a large proportion of patients. Here, we investigated the feasibility of this combination by analyzing the effect of vemurafenib on the functionality of nDCs. METHODS: Plasmacytoid DCs (pDCs) and myeloid DCs (mDCs) were isolated from PBMCs obtained from buffy coats from healthy volunteers or vemurafenib-treated melanoma patients. Maturation of pDCs, mDCs and immature monocyte-derived DCs was induced by R848 in the presence or absence of vemurafenib and analyzed by FACS. Cytokine production and T cell proliferation induced by mature DCs were analyzed. RESULTS: Vemurafenib inhibited maturation and cytokine production of highly purified nDCs of healthy volunteers resulting in diminished allogeneic T cell proliferation. This deleterious effect of vemurafenib on nDC functionality was absent when total PBMCs were exposed to vemurafenib. In patients receiving vemurafenib, nDC functionality and T cell allostimulatory capacity were unaffected. CONCLUSION: Although vemurafenib inhibited the functionality of purified nDC of healthy volunteers, this effect was not observed when nDCs were matured in the complete PBMC fraction. This might have been caused by increased vemurafenib uptake in absence of other cell types. In accordance, nDCs isolated from patients on active vemurafenib treatment showed no negative effects. In conclusion, our results pave the way for a combinatorial treatment strategy and, we propose that combining vemurafenib with nDC vaccination represent a powerful opportunity that deserves more investigation in the clinic.


Asunto(s)
Células Dendríticas/inmunología , Células Mieloides/inmunología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Vacunación , Presentación de Antígeno/efectos de los fármacos , Antígenos de Neoplasias/inmunología , Disponibilidad Biológica , Diferenciación Celular/efectos de los fármacos , Separación Celular , Citocinas/metabolismo , Células Dendríticas/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Humanos , Indoles/sangre , Indoles/farmacología , Activación de Linfocitos/efectos de los fármacos , Melanoma/sangre , Melanoma/patología , Células Mieloides/efectos de los fármacos , Proteínas Proto-Oncogénicas B-raf/metabolismo , Piridonas/farmacología , Pirimidinonas/farmacología , Sulfonamidas/sangre , Sulfonamidas/farmacología , Vemurafenib
10.
Gynecol Oncol ; 137(2): 335-42, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25727651

RESUMEN

Ovarian cancer is a devastating disease with a high relapse rate. Due to a mostly asymptomatic early stage and lack of early diagnostic tools, the disease is usually diagnosed in a late stage. Surgery and chemotherapy with taxanes and platinum compounds are very effective in reducing tumor burden. However, relapses occur frequently and there is a lack of credible second-line options. Therefore, new treatment modalities are eagerly awaited. The presence and influx of immune cells in the ovarian cancer tumor microenvironment are correlated with survival. High numbers of infiltrating T cells correlate with improved progression free and overall survival, while the presence of regulatory T cells and expression of T cell inhibitory molecules is correlated with a poor prognosis. These data indicate that immunotherapy, especially cell-based immunotherapy could be a promising novel addition to the treatment of ovarian cancer. Here, we review the available data on the immune contexture surrounding ovarian cancer and discuss novel strategies and targets for immunotherapy in ovarian cancer. In the end the addition of immunotherapy to existing therapeutic options could lead to a great improvement in the outcome of ovarian cancer, especially when targeting cancer stem cells.


Asunto(s)
Inmunoterapia/métodos , Células Madre Neoplásicas/inmunología , Neoplasias Ováricas/terapia , Femenino , Humanos , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/patología , Células Madre Neoplásicas/patología , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología
11.
Clin Cancer Res ; 20(11): 2831-7, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24879823

RESUMEN

The platinum-based drugs cisplatin, carboplatin, and oxaliplatin belong to the most widely used chemotherapeutics in oncology, showing clinical efficacy against many solid tumors. Their main mechanism of action is believed to be the induction of cancer cell apoptosis as a response to their covalent binding to DNA. In recent years, this picture has increased in complexity, based on studies indicating that cellular molecules other than DNA may potentially act as targets, and that part of the antitumor effects of platinum drugs occurs through modulation of the immune system. These immunogenic effects include modulation of STAT signaling; induction of an immunogenic type of cancer cell death through exposure of calreticulin and release of ATP and high-mobility group protein box-1 (HMGB-1); and enhancement of the effector immune response through modulation of programmed death receptor 1-ligand and mannose-6-phosphate receptor expression. Both basic and clinical studies indicate that at least part of the antitumor efficacy of platinum chemotherapeutics may be due to immune potentiating mechanisms. Clinical studies exploiting this novel mechanism of action of these old cancer drugs have been initiated. Here, we review the literature on the immunogenic effects of platinum, summarize the clinical advances using platinum as a cytotoxic compound with immune adjuvant properties, and discuss the limitations to these studies and the gaps in our understanding of the immunologic effects of these drugs. Clin Cancer Res; 20(11); 2831-7. ©2014 AACR.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Compuestos de Platino/farmacología , Humanos
12.
J Virol ; 88(6): 3369-78, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24390337

RESUMEN

UNLABELLED: RIG-I-like receptors (RLRs) MDA5 and RIG-I are key players in the innate antiviral response. Upon recognition of viral RNA, they interact with MAVS, eventually inducing type I interferon production. The interferon induction pathway is commonly targeted by viruses. How enteroviruses suppress interferon production is incompletely understood. MDA5 has been suggested to undergo caspase- and proteasome-mediated degradation during poliovirus infection. Additionally, MAVS is reported to be cleaved during infection with coxsackievirus B3 (CVB3) by the CVB3 proteinase 3C(pro), whereas MAVS cleavage by enterovirus 71 has been attributed to 2A(pro). As yet, a detailed examination of the RLR pathway as a whole during any enterovirus infection is lacking. We performed a comprehensive analysis of crucial factors of the RLR pathway, including MDA5, RIG-I, LGP2, MAVS, TBK1, and IRF3, during infection of CVB3, a human enterovirus B (HEV-B) species member. We show that CVB3 inhibits the RLR pathway upstream of TBK1 activation, as demonstrated by limited phosphorylation of TBK1 and a lack of IRF3 phosphorylation. Furthermore, we show that MDA5, MAVS, and RIG-I all undergo proteolytic degradation in CVB3-infected cells through a caspase- and proteasome-independent manner. We convincingly show that MDA5 and MAVS cleavages are both mediated by CVB3 2A(pro), while RIG-I is cleaved by 3C(pro). Moreover, we show that proteinases 2A(pro) and 3C(pro) of poliovirus (HEV-C) and enterovirus 71 (HEV-A) exert the same functions. This study identifies a critical role of 2A(pro) by cleaving MDA5 and MAVS and shows that enteroviruses use a common strategy to counteract the interferon response in infected cells. IMPORTANCE: Human enteroviruses (HEVs) are important pathogens that cause a variety of diseases in humans, including poliomyelitis, hand, foot, and mouth disease, viral meningitis, cardiomyopathy, and more. Like many other viruses, enteroviruses target the host immune pathways to gain replication advantage. The MDA5/MAVS pathway is responsible for recognizing enterovirus infections in the host cell and leads to expression of type I interferons (IFN-I), crucial antiviral signaling molecules. Here we show that three species of HEVs all employ the viral proteinase 2A (2A(pro)) to proteolytically target MDA5 and MAVS, leading to an efficient blockade upstream of IFN-I transcription. These observations suggest that MDA5/MAVS antagonization is an evolutionarily conserved and beneficial mechanism of enteroviruses. Understanding the molecular mechanisms of enterovirus immune evasion strategies will help to develop countermeasures to control infections with these viruses in the future.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Cisteína Endopeptidasas/metabolismo , ARN Helicasas DEAD-box/metabolismo , Enterovirus Humano B/enzimología , Infecciones por Enterovirus/metabolismo , Proteínas Virales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Cisteína Endopeptidasas/genética , ARN Helicasas DEAD-box/genética , Enterovirus Humano B/genética , Enterovirus Humano B/fisiología , Infecciones por Enterovirus/enzimología , Infecciones por Enterovirus/genética , Infecciones por Enterovirus/virología , Interacciones Huésped-Patógeno , Humanos , Helicasa Inducida por Interferón IFIH1 , Fosforilación , Proteolisis , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Transducción de Señal , Proteínas Virales/genética
13.
Crit Rev Immunol ; 34(6): 517-36, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25597314

RESUMEN

Dendritic cells (DCs) are key players in the induction of immune responses. Adoptive transfer of autologous mature DCs loaded with tumor-associated antigens is a promising therapy for the treatment of immunogenic tumors. For a long time, its therapeutic activity was thought to depend solely on the induction of tumor-specific CD8+ and CD4+ T cell responses. More recently, DCs were shown to bidirectionally interact with innate and innate-like immune cells, including natural killer (NK), invariant natural killer T (iNKT), and γδ T cells. These effector cells can amplify responses induced by DCs via several mechanisms, including induction of DC maturation and conventional T cell priming. In addition, NK, iNKT, and γδ T cells possess cytolytic activity and can act directly on tumor cells. Therapeutic strategies targeting these innate and innate-like immune cells hence hold potential to improve current DC vaccination protocols.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Células Asesinas Naturales/inmunología , Neoplasias/prevención & control , Linfocitos T Citotóxicos/inmunología , Traslado Adoptivo , Animales , Antígenos de Neoplasias/genética , Linfocitos T CD4-Positivos/citología , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Comunicación Celular/inmunología , Células Dendríticas/citología , Células Dendríticas/trasplante , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunidad Innata , Células Asesinas Naturales/citología , Ratones , Neoplasias/inmunología , Neoplasias/patología , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Transducción de Señal , Linfocitos T Citotóxicos/citología , Vacunación
14.
Front Immunol ; 4: 417, 2013 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-24348481

RESUMEN

The aim of therapeutic dendritic cell (DC) vaccines in cancer immunotherapy is to activate cytotoxic T cells to recognize and attack the tumor. T cell activation requires the interaction of the T cell receptor with a cognate major-histocompatibility complex-peptide complex. Although initiated by antigen engagement, it is the complex balance between co-stimulatory and co-inhibitory signals on DCs that results in T cell activation or tolerance. Even when already activated, tumor-specific T cells can be neutralized by the expression of co-inhibitory molecules on tumor cells. These and other immunosuppressive cues in the tumor microenvironment are major factors currently hampering the application of DC vaccination. In this review, we discuss recent data regarding the essential and complex role of co-inhibitory molecules in regulating the immune response within the tumor microenvironment. In particular, possible therapeutic intervention strategies aimed at reversing or neutralizing suppressive networks within the tumor microenvironment will be emphasized. Importantly, blocking co-inhibitory molecule signaling, often referred to as immune checkpoint blockade, does not necessarily lead to an effective activation of tumor-specific T cells. Therefore, combination of checkpoint blockade with other immune potentiating therapeutic strategies, such as DC vaccination, might serve as a synergistic combination, capable of reversing effector T cells immunosuppression while at the same time increasing the efficacy of T cell-mediated immunotherapies. This will ultimately result in long-term anti-tumor immunity.

15.
J Clin Invest ; 123(2): 917-27, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23348739

RESUMEN

Topical application of coal tar is one of the oldest therapies for atopic dermatitis (AD), a T helper 2 (Th2) lymphocyte-mediated skin disease associated with loss-of-function mutations in the skin barrier gene, filaggrin (FLG). Despite its longstanding clinical use and efficacy, the molecular mechanism of coal tar therapy is unknown. Using organotypic skin models with primary keratinocytes from AD patients and controls, we found that coal tar activated the aryl hydrocarbon receptor (AHR), resulting in induction of epidermal differentiation. AHR knockdown by siRNA completely abrogated this effect. Coal tar restored filaggrin expression in FLG-haploinsufficient keratinocytes to wild-type levels, and counteracted Th2 cytokine-mediated downregulation of skin barrier proteins. In AD patients, coal tar completely restored expression of major skin barrier proteins, including filaggrin. Using organotypic skin models stimulated with Th2 cytokines IL-4 and IL-13, we found coal tar to diminish spongiosis, apoptosis, and CCL26 expression, all AD hallmarks. Coal tar interfered with Th2 cytokine signaling via dephosphorylation of STAT6, most likely due to AHR-regulated activation of the NRF2 antioxidative stress pathway. The therapeutic effect of AHR activation herein described opens a new avenue to reconsider AHR as a pharmacological target and could lead to the development of mechanism-based drugs for AD.


Asunto(s)
Alquitrán/administración & dosificación , Dermatitis Atópica/tratamiento farmacológico , Dermatitis Atópica/fisiopatología , Receptores de Hidrocarburo de Aril/efectos de los fármacos , Receptores de Hidrocarburo de Aril/fisiología , Administración Tópica , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Citocinas/metabolismo , Dermatitis Atópica/inmunología , Dermatitis Atópica/patología , Proteínas Filagrina , Humanos , Proteínas de Filamentos Intermediarios/genética , Proteínas de Filamentos Intermediarios/metabolismo , Queratinocitos/efectos de los fármacos , Queratinocitos/patología , Queratinocitos/fisiología , Modelos Biológicos , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo/efectos de los fármacos , ARN Interferente Pequeño/genética , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , Receptores de Hidrocarburo de Aril/genética , Transducción de Señal/efectos de los fármacos , Células Th2/inmunología , Regulación hacia Arriba/efectos de los fármacos
16.
Cell Rep ; 2(5): 1187-96, 2012 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-23142662

RESUMEN

RIG-I and MDA5 are cytosolic RNA sensors that play a critical role in innate antiviral responses. Major advances have been made in identifying RIG-I ligands, but our knowledge of the ligands for MDA5 remains restricted to data from transfection experiments mostly using poly(I:C), a synthetic dsRNA mimic. Here, we dissected the IFN-α/ß-stimulatory activity of different viral RNA species produced during picornavirus infection, both by RNA transfection and in infected cells in which specific steps of viral RNA replication were inhibited. Our results show that the incoming genomic plus-strand RNA does not activate MDA5, but minus-strand RNA synthesis and production of the 7.5 kbp replicative form trigger a strong IFN-α/ß response. IFN-α/ß production does not rely on plus-strand RNA synthesis and thus generation of the partially double-stranded replicative intermediate. This study reports MDA5 activation by a natural RNA ligand under physiological conditions.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , ARN Bicatenario/metabolismo , ARN Viral/metabolismo , Animales , Línea Celular , ARN Helicasas DEAD-box/química , Células HeLa , Caballos , Humanos , Helicasa Inducida por Interferón IFIH1 , Interferón-alfa/metabolismo , Interferón beta/genética , Interferón beta/metabolismo , Ratones , Picornaviridae/genética , Poli I-C/farmacología , ARN Bicatenario/genética , ARN Mensajero/metabolismo , Transfección , Regulación hacia Arriba/efectos de los fármacos
17.
Oncoimmunology ; 1(2): 234-236, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22720254

RESUMEN

Platinum-based anticancer drugs enhance the immunostimulatory potential of DCs and decrease the immunosuppressive capacity of tumor cells. This immunomodulatory ability is based on the inhibition of STAT6-mediated expression of co-inhibitory molecule PD-L2 and opens up the possibility of using these drugs in combination with other immunostimulatory compounds.

18.
Clin Dev Immunol ; 2012: 656340, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22611421

RESUMEN

Inhibitory molecules of the B7/CD28 family play a key role in the induction of immune tolerance in the tumor microenvironment. The programmed death-1 receptor (PD-1), with its ligands PD-L1 and PD-L2, constitutes an important member of these inhibitory pathways. The relevance of the PD-1/PD-L1 pathway in cancer has been extensively studied and therapeutic approaches targeting PD-1 and PD-L1 have been developed and are undergoing human clinical testing. However, PD-L2 has not received as much attention and its role in modulating tumor immunity is less clear. Here, we review the literature on the immunobiology of PD-L2, particularly on its possible roles in cancer-induced immune suppression and we discuss the results of recent studies targeting PD-L2 in cancer.


Asunto(s)
Terapia de Inmunosupresión , Neoplasias/inmunología , Proteína 2 Ligando de Muerte Celular Programada 1/inmunología , Animales , Antineoplásicos/uso terapéutico , Ensayos Clínicos como Asunto , Humanos , Ratones , Terapia Molecular Dirigida/tendencias , Neoplasias/tratamiento farmacológico , Escape del Tumor , Microambiente Tumoral
19.
Cancer Immunol Immunother ; 61(7): 1101-11, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22193989

RESUMEN

It has become evident that the tumor microenvironment plays a pivotal role in the maintenance of cancerous growth. One of the acquired functions of the tumor microenvironment is the suppression of immune responses. Indeed, blocking the inhibitory pathways operational in the microenvironment results in enhanced T-cell-dependent, anti-tumor immunity. Chemotherapeutic drugs not only directly kill tumor cells but also shape the tumor microenvironment and potentiate anti-tumor immunity. Here, we demonstrate that the chemotherapeutic compound oxaliplatin acts as a double-edged sword. Besides killing tumor cells, oxaliplatin bolsters immunosuppressive pathways, resulting in decreased activation of T cells by human plasmacytoid dendritic cells (pDCs). Exposure to oxaliplatin markedly increased expression of the T-cell inhibitory molecule programmed death receptor-ligand 1 (PD-L1) on human pDCs and also TLR9-induced IFNα secretion. Furthermore, oxaliplatin decreased TLR-induced STAT1 and STAT3 expression, and NF-κB-mediated responses. The oxaliplatin induced upregulation of PD-L1 and downregulation of costimulatory molecules CD80 and CD86 resulted in decreased T-cell proliferation. Our results demonstrate that platinum-based anticancer drugs adapt TLR-induced signaling in human pDCs and myeloid DCs (mDCs), thereby downgrading their immunostimulatory potential.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Compuestos Organoplatinos/farmacología , Antineoplásicos/farmacología , Antígeno B7-H1/inmunología , Diferenciación Celular , Procesos de Crecimiento Celular/efectos de los fármacos , Procesos de Crecimiento Celular/inmunología , Células Dendríticas/inmunología , Humanos , Interferón Tipo I/inmunología , Interleucina-6/inmunología , Activación de Linfocitos/efectos de los fármacos , Oxaliplatino , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT3/inmunología , Receptor Toll-Like 9/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Factor de Necrosis Tumoral alfa/inmunología
20.
J Clin Invest ; 121(8): 3100-8, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21765211

RESUMEN

Tumor microenvironments feature immune inhibitory mechanisms that prevent T cells from generating effective antitumor immune responses. Therapeutic interventions aimed at disrupting these inhibitory mechanisms have been shown to enhance antitumor immunity, but they lack direct cytotoxic effects. Here, we investigated the effect of cytotoxic cancer chemotherapeutics on immune inhibitory pathways. We observed that exposure to platinum-based chemotherapeutics markedly reduced expression of the T cell inhibitory molecule programmed death receptor-ligand 2 (PD-L2) on both human DCs and human tumor cells. Downregulation of PD-L2 resulted in enhanced antigen-specific proliferation and Th1 cytokine secretion as well as enhanced recognition of tumor cells by T cells. Further analysis revealed that STAT6 controlled downregulation of PD-L2. Consistent with these data, patients with STAT6-expressing head and neck cancer displayed enhanced recurrence-free survival upon treatment with cisplatin-based chemoradiation compared with patients with STAT6-negative tumors, demonstrating the clinical relevance of platinum-induced STAT6 modulation. We therefore conclude that platinum-based anticancer drugs can enhance the immunostimulatory potential of DCs and decrease the immunosuppressive capability of tumor cells. This dual action of platinum compounds may extend their therapeutic application in cancer patients and provides a rationale for their use in combination with immunostimulatory compounds.


Asunto(s)
Cisplatino/farmacología , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Factor de Transcripción STAT6/metabolismo , Animales , Antineoplásicos/farmacología , Proliferación Celular , Células Dendríticas/citología , Supervivencia sin Enfermedad , Regulación hacia Abajo , Humanos , Sistema Inmunológico , Ratones , Ratones Endogámicos BALB C , Fosforilación , Linfocitos T/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA