Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 43(4): 114062, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38588339

RESUMEN

The role of T cell receptor (TCR) diversity in infectious disease susceptibility is not well understood. We use a systems immunology approach on three cohorts of herpes zoster (HZ) patients and controls to investigate whether TCR diversity against varicella-zoster virus (VZV) influences the risk of HZ. We show that CD4+ T cell TCR diversity against VZV glycoprotein E (gE) and immediate early 63 protein (IE63) after 1-week culture is more restricted in HZ patients. Single-cell RNA and TCR sequencing of VZV-specific T cells shows that T cell activation pathways are significantly decreased after stimulation with VZV peptides in convalescent HZ patients. TCR clustering indicates that TCRs from HZ patients co-cluster more often together than TCRs from controls. Collectively, our results suggest that not only lower VZV-specific TCR diversity but also reduced functional TCR affinity for VZV-specific proteins in HZ patients leads to lower T cell activation and consequently affects the susceptibility for viral reactivation.


Asunto(s)
Herpes Zóster , Herpesvirus Humano 3 , Activación de Linfocitos , Receptores de Antígenos de Linfocitos T , Humanos , Herpes Zóster/inmunología , Herpes Zóster/virología , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Activación de Linfocitos/inmunología , Herpesvirus Humano 3/inmunología , Femenino , Persona de Mediana Edad , Masculino , Linfocitos T CD4-Positivos/inmunología , Anciano , Adulto , Epítopos de Linfocito T/inmunología
2.
Front Immunol ; 9: 2503, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30464762

RESUMEN

Genetic engineering of T cells with tumor specific T-cell receptors (TCR) is a promising strategy to redirect their specificity against cancer cells in adoptive T cell therapy protocols. Most studies are exploiting integrating retro- or lentiviral vectors to permanently introduce the therapeutic TCR, which can pose serious safety issues when treatment-related toxicities would occur. Therefore, we developed a versatile, non-genotoxic transfection method for human unstimulated CD8+ T cells. We describe an optimized double sequential electroporation platform whereby Dicer-substrate small interfering RNAs (DsiRNA) are first introduced to suppress endogenous TCR α and ß expression, followed by electroporation with DsiRNA-resistant tumor-specific TCR mRNA. We demonstrate that double sequential electroporation of human primary unstimulated T cells with DsiRNA and TCR mRNA leads to unprecedented levels of transgene TCR expression due to a strongly reduced degree of TCR mispairing. Importantly, superior transgenic TCR expression boosts epitope-specific CD8+ T cell activation and killing activity. Altogether, DsiRNA and TCR mRNA double sequential electroporation is a rapid, non-integrating and highly efficient approach with an enhanced biosafety profile to engineer T cells with antigen-specific TCRs for use in early phase clinical trials.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Ingeniería Genética/métodos , Inmunoterapia Adoptiva/métodos , Neoplasias/terapia , ARN/genética , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/trasplante , Citotoxicidad Inmunológica , Electroporación , Epítopos de Linfocito T/inmunología , Vectores Genéticos , Humanos , Neoplasias/inmunología , ARN Interferente Pequeño/genética , Ribonucleasa III/metabolismo , Especificidad del Receptor de Antígeno de Linfocitos T
3.
Oncotarget ; 9(45): 27797-27808, 2018 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-29963238

RESUMEN

Blockade of programmed cell death protein 1 (PD-1) immune checkpoint receptor signaling is an established standard treatment for many types of cancer and indications are expanding. Successful clinical trials using monoclonal antibodies targeting PD-1 signaling have boosted preclinical research, encouraging development of novel therapeutics. Standardized assays to evaluate their bioactivity, however, remain restricted. The robust bioassays available all lack antigen-specificity. Here, we developed an antigen-specific, short-term and high-throughput T cell assay with versatile readout possibilities. A genetically modified T cell receptor (TCR)-deficient T cell line was stably transduced with PD-1. Transfection with messenger RNA encoding a TCR of interest and subsequent overnight stimulation with antigen-presenting cells, results in eGFP-positive and granzyme B-producing T cells for single cell or bulk analysis. Control antigen-presenting cells induced reproducible high antigen-specific eGFP and granzyme B expression. Upon PD-1 interaction, ligand-positive antigen-presenting immune or tumor cells elicited significantly lower eGFP and granzyme B expression, which could be restored by anti-PD-(L)1 blocking antibodies. This convenient cell-based assay shows a valuable tool for translational and clinical research on antigen-specific checkpoint-targeted therapy approaches.

4.
Front Immunol ; 9: 394, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29599770

RESUMEN

Two decades of clinical cancer research with dendritic cell (DC)-based vaccination have proved that this type of personalized medicine is safe and has the capacity to improve survival, but monotherapy is unlikely to cure the cancer. Designed to empower the patient's antitumor immunity, huge research efforts are set to improve the efficacy of next-generation DC vaccines and to find synergistic combinations with existing cancer therapies. Immune checkpoint approaches, aiming to breach immune suppression and evasion to reinforce antitumor immunity, have been a revelation in the immunotherapy field. Early success of therapeutic antibodies blocking the programmed death-1 (PD-1) pathway has sparked the development of novel inhibitors and combination therapies. Hence, merging immunoregulatory tumor-specific DC strategies with PD-1-targeted approaches is a promising path to explore. In this review, we focus on the role of PD-1-signaling in DC-mediated antitumor immunity. In the quest of exploiting the full potential of DC therapy, different strategies to leverage DC immunopotency by impeding PD-1-mediated immune regulation are discussed, including the most advanced research on targeted therapeutic antibodies, lessons learned from chemotherapy-induced immune activation, and more recent developments with soluble molecules and gene-silencing techniques. An overview of DC/PD-1 immunotherapy combinations that are currently under preclinical and clinical investigation substantiates the clinical potential of such combination strategies.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Vacunas contra el Cáncer/inmunología , Células Dendríticas/trasplante , Inmunoterapia/métodos , Neoplasias/terapia , Animales , Terapia Combinada , Células Dendríticas/inmunología , Evaluación Preclínica de Medicamentos , Humanos , Neoplasias/inmunología , Receptor de Muerte Celular Programada 1/inmunología
5.
Oncoimmunology ; 7(3): e1407899, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29399410

RESUMEN

Prognosis of glioblastoma remains dismal, underscoring the need for novel therapies. Immunotherapy is generating promising results, but requires combination strategies to unlock its full potential. We investigated the immunomodulatory capacities of poly(I:C) on primary human glioblastoma cells and its combinatorial potential with programmed death ligand (PD-L) blockade. In our experiments, poly(I:C) stimulated expression of both PD-L1 and PD-L2 on glioblastoma cells, and a pro-inflammatory secretome, including type I interferons (IFN) and chemokines CXCL9, CXCL10, CCL4 and CCL5. IFN-ß was partially responsible for the elevated PD-1 ligand expression on these cells. Moreover, real-time PCR and chloroquine-mediated blocking experiments indicated that poly(I:C) triggered Toll-like receptor 3 to elicit its effect. Cocultures of poly(I:C)-treated glioblastoma cells with peripheral blood mononuclear cells enhanced lymphocytic activation (CD69, IFN-γ) and cytotoxic capacity (CD107a, granzyme B). Additional PD-L1 blockade further propagated immune activation. Besides activating immunity, poly(I:C)-treated glioblastoma cells also doubled the attraction of CD8+ T cells, and to a lesser extent CD4+ T cells, via a mechanism which included CXCR3 and CCR5 ligands. Our results indicate that by triggering glioblastoma cells, poly(I:C) primes the tumor microenvironment for an immune response. Secreted cytokines allow for immune activation while chemokines attract CD8+ T cells to the front, which are postulated as a prerequisite for effective PD-1/PD-L1 blockade. Accordingly, additional blockade of the concurrently elevated tumoral PD-L1 further reinforces the immune activation. In conclusion, our data proposes poly(I:C) treatment combined with PD-L1 blockade to invigorate the immune checkpoint inhibition response in glioblastoma.

6.
Immunogenetics ; 70(6): 363-372, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29196796

RESUMEN

Around 30% of individuals will develop herpes zoster (HZ), caused by the varicella zoster virus (VZV), during their life. While several risk factors for HZ, such as immunosuppressive therapy, are well known, the genetic and molecular components that determine the risk of otherwise healthy individuals to develop HZ are still poorly understood. We created a computational model for the Human Leukocyte Antigen (HLA-A, -B, and -C) presentation capacity of peptides derived from the VZV Immediate Early 62 (IE62) protein. This model could then be applied to a HZ cohort with known HLA molecules. We found that HLA-A molecules with poor VZV IE62 presentation capabilities were more common in a cohort of 50 individuals with a history of HZ compared to a nationwide control group, which equated to a HZ risk increase of 60%. This tendency was most pronounced for cases of HZ at a young age, where other risk factors are less prevalent. These findings provide new molecular insights into the development of HZ and reveal a genetic predisposition in those individuals most at risk to develop HZ.


Asunto(s)
Antígenos HLA-A/inmunología , Herpes Zóster/inmunología , Herpesvirus Humano 3/inmunología , Proteínas Inmediatas-Precoces/inmunología , Transactivadores/inmunología , Proteínas del Envoltorio Viral/inmunología , Adulto , Anciano , Bélgica/epidemiología , Varicela/inmunología , Varicela/virología , Femenino , Predisposición Genética a la Enfermedad , Herpes Zóster/epidemiología , Herpes Zóster/genética , Humanos , Proteínas Inmediatas-Precoces/genética , Masculino , Persona de Mediana Edad , Modelos Inmunológicos , Factores de Riesgo , Transactivadores/genética , Proteínas del Envoltorio Viral/genética
7.
Ind Eng Chem Res ; 56(45): 13423-13433, 2017 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-29170600

RESUMEN

ZnCl2 hydrate, the main molten salt used in biomass conversion, combined with low concentration HCl is an excellent solvent for the dissolution and hydrolysis of the carbohydrates present in lignocellulosic biomass. The most recalcitrant carbohydrate, cellulose, is dissolved in a residence time less than 1 h under mild conditions without significant degradation. This technology is referred to as BIOeCON-solvent technology. Separation of the sugars from the solution is the main challenge. The earlier conclusion regarding the potential of zeolite beta for selective adsorption has been used as the basis of a scale-up study. The technology of choice is continuous chromatographic separation (e.g., simulated moving bed, SMB). The sugar monomers are separated from the sugar oligomers, allowing the production of monosugars at high yield, using water as an eluent. Results of a pilot plant study are presented showing a stable operation at high selectivity. Several process designs are discussed, and the techno-economic performance of the BIOeCON-solvent technology is demonstrated by comparison with the state-of-the-art technology of NREL (National Renewable Energy Laboratory), which is based on enzymatic conversion of cellulose. It is concluded that the BIOeCON-solvent technology is technically and economically viable and is competitive to the NREL process. Because the BIOeCON-solvent process is in an early stage of development and far from fully optimized, it has the potential to outperform the existing processes.

8.
Oncotarget ; 8(34): 56968-56979, 2017 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-28915646

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is the 4th leading cause of cancer-related death in Western countries with a 5-year survival rate below 5%. One of the hallmarks of this cancer is the strong desmoplastic reaction within the tumor microenvironment (TME), orchestrated by activated pancreatic stellate cells (PSC). This results in a functional and mechanical shield which causes resistance to conventional therapies. Aiming to overcome this resistance by tackling the stromal shield, we assessed for the first time the capacity of IL-15 stimulated natural killer (NK) cells to kill PSC and pancreatic cancer cells (PCC). The potency of IL-15 to promote NK cell-mediated killing was evaluated phenotypically and functionally. In addition, NK cell and immune checkpoint ligands on PSC were charted. We demonstrate that IL-15 activated NK cells kill both PCC and PSC lines (range 9-35% and 20-50%, respectively) in a contact-dependent manner and significantly higher as compared to resting NK cells. Improved killing of these pancreatic cell lines is, at least partly, dependent on IL-15 induced upregulation of TIM-3 and NKG2D. Furthermore, we confirm significant killing of primary PSC by IL-15 activated NK cells in an ex vivo autologous system. Screening for potential targets for immunotherapeutic strategies, we demonstrate surface expression of both inhibitory (PD-L1, PD-L2) and activating (MICA/B, ULBPs and Galectin-9) ligands on primary PSC. These data underscore the therapeutic potential of IL-15 to promote NK cell-mediated cytotoxicity as a treatment of pancreatic cancer and provide promising future targets to tackle remaining PSC.

9.
Cancer Immunol Res ; 5(8): 710-715, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28637876

RESUMEN

Although allogeneic stem cell transplantation (allo-SCT) can elicit graft-versus-tumor (GVT) immunity, patients often relapse due to residual tumor cells. As essential orchestrators of the immune system, vaccination with dendritic cells (DC) is an appealing strategy to boost the GVT response. Nevertheless, durable clinical responses after DC vaccination are still limited, stressing the need to improve current DC vaccines. Aiming to empower DC potency, we engineered monocyte-derived DCs to deprive them of ligands for the immune checkpoint regulated by programmed death 1 (PD-1). We also equipped them with interleukin (IL)-15 "transpresentation" skills. Transfection with short interfering (si)RNA targeting the PD-1 ligands PD-L1 and PD-L2, in combination with IL15 and IL15Rα mRNA, preserved their mature DC profile and rendered the DCs superior in inducing T-cell proliferation and IFNγ and TNFα production. Translated into an ex vivo hematological disease setting, DCs deprived of PD-1 ligands (PD-L), equipped with IL15/IL15Rα expression, or most effectively, both, induced superior expansion of minor histocompatibility antigen-specific CD8+ T cells from transplanted cancer patients. These data support the combinatorial approach of in situ suppression of the PD-L inhibitory checkpoints with DC-mediated IL15 transpresentation to promote antigen-specific T-cell responses and, ultimately, contribute to GVT immunity. Cancer Immunol Res; 5(8); 710-5. ©2017 AACR.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Células Dendríticas/trasplante , Interleucina-15/genética , Receptor de Muerte Celular Programada 1/genética , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Efecto Injerto vs Tumor/efectos de los fármacos , Efecto Injerto vs Tumor/inmunología , Humanos , Interleucina-15/antagonistas & inhibidores , Monocitos/inmunología , Monocitos/trasplante , Proteína 2 Ligando de Muerte Celular Programada 1/genética , Proteína 2 Ligando de Muerte Celular Programada 1/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , ARN Interferente Pequeño/genética , Trasplante de Células Madre , Transfección , Trasplante Homólogo , Vacunación
10.
Oncotarget ; 8(8): 13652-13665, 2017 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-28099143

RESUMEN

Success of dendritic cell (DC) therapy in treating malignancies is depending on the DC capacity to attract immune effector cells, considering their reciprocal crosstalk is partially regulated by cell-contact-dependent mechanisms. Although critical for therapeutic efficacy, immune cell recruitment is a largely overlooked aspect regarding optimization of DC vaccination. In this paper we have made a head-to-head comparison of interleukin (IL)-15-cultured DCs and conventional IL-4-cultured DCs with regard to their proficiency in the recruitment of (innate) immune effector cells. Here, we demonstrate that IL-4 DCs are suboptimal in attracting effector lymphocytes, while IL15 DCs provide a favorable chemokine milieu for recruiting CD8+ T cells, natural killer (NK) cells and gamma delta (γδ) T cells. Gene expression analysis revealed that IL-15 DCs exhibit a high expression of chemokines involved in antitumor immune effector cell attraction, while IL-4 DCs display a more immunoregulatory profile characterized by the expression of Th2 and regulatory T cell-attracting chemokines. This is confirmed by functional data indicating an enhanced recruitment of granzyme B+ effector lymphocytes by IL-15 DCs, as compared to IL-4 DCs, and subsequent superior killing of tumor cells by the migrated lymphocytes. Elevated CCL4 gene expression in IL-15 DCs and lowered CCR5 expression on both migrated γδ T cells and NK cells, led to validation of increased CCL4 secretion by IL15 DCs. Moreover, neutralization of CCR5 prior to migration resulted in an important inhibition of γδ T cell and NK cell recruitment by IL-15 DCs. These findings further underscore the strong immunotherapeutic potential of IL-15 DCs.


Asunto(s)
Células Dendríticas/inmunología , Inmunoterapia Adoptiva/métodos , Interleucina-15/inmunología , Células Asesinas Naturales/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Movimiento Celular/inmunología , Quimiocinas/genética , Quimiocinas/inmunología , Expresión Génica , Humanos , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Proteínas de Transporte Vesicular/inmunología
11.
Front Immunol ; 8: 1964, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29403473

RESUMEN

The use of tolerance-inducing dendritic cells (tolDCs) has been proven to be safe and well tolerated in the treatment of autoimmune diseases. Nevertheless, several challenges remain, including finding ways to facilitate the migration of cell therapeutic products to lymph nodes, and the site of inflammation. In the treatment of neuroinflammatory diseases, such as multiple sclerosis (MS), the blood-brain barrier (BBB) represents a major obstacle to the delivery of therapeutic agents to the inflamed central nervous system (CNS). As it was previously demonstrated that C-C chemokine receptor 5 (CCR5) may be involved in inflammatory migration of DCs, the aim of this study was to investigate CCR5-driven migration of tolDCs. Only a minority of in vitro generated vitamin D3 (vitD3)-treated tolDCs expressed the inflammatory chemokine receptor CCR5. Thus, messenger RNA (mRNA) encoding CCR5 was introduced by means of electroporation (EP). After mRNA EP, tolDCs transiently displayed increased levels of CCR5 protein expression. Accordingly, the capacity of mRNA electroporated tolDCs to transmigrate toward a chemokine gradient in an in vitro model of the BBB improved significantly. Neither the tolerogenic phenotype nor the T cell-stimulatory function of tolDCs was affected by mRNA EP. EP of tolDCs with mRNA encoding CCR5 enabled these cells to migrate to inflammatory sites. The approach used herein has important implications for the treatment of MS. Using this approach, tolDCs actively shuttle across the BBB, allowing in situ down-modulation of autoimmune responses in the CNS.

12.
Pharmacol Ther ; 170: 73-79, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27777088

RESUMEN

Interleukin (IL)-15 as a stand-alone therapy can activate the antitumor functions of immune effector cells resulting in significant tumor regression. Interestingly, combining IL-15 with the α-moiety of its receptor (IL-15Rα), also called IL-15 transpresentation, increases the in vivo half-life of IL-15 and enhances binding of IL-15 with cells expressing the IL-15Rßγ, such as NK cells and CD8+ T cells. These features enlarge the signal transmission of IL-15, resulting in improved proliferation and antitumor activities of both NK cells and CD8+ T cells, eventually leading to enhanced killing of tumor cells. In this review, we discuss the antitumor strategies in which this IL-15 transpresentation mechanism is implemented, that are currently under preclinical investigation. Furthermore, we give an overview of the studies in which the IL-15/IL-15Rα complexes are combined with other antitumor therapies. The promising results in these preclinical studies have incited several clinical trials to test the safety and efficacy of IL-15 transpresentation strategies to treat both hematological and advanced solid tumors.


Asunto(s)
Subunidad alfa del Receptor de Interleucina-15/inmunología , Interleucina-15/administración & dosificación , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/inmunología , Linfocitos T CD8-positivos/inmunología , Neoplasias Hematológicas/tratamiento farmacológico , Neoplasias Hematológicas/inmunología , Humanos , Interleucina-15/inmunología , Células Asesinas Naturales/inmunología , Neoplasias/inmunología , Neoplasias/patología
13.
J Hematol Oncol ; 9(1): 101, 2016 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-27686372

RESUMEN

BACKGROUND: Adoptive immunotherapy is gaining momentum to fight malignancies, whereby γδ T cells have received recent attention as an alternative cell source as to natural killer cells and αß T cells. The advent of γδ T cells is largely due to their ability to recognize and target tumor cells using both innate characteristic and T cell receptor (TCR)-mediated mechanisms, their capacity to enhance the generation of antigen-specific T cell responses, and their potential to be used in an autologous or allogeneic setting. METHODS: In this study, we explored the beneficial effect of the immunostimulatory cytokine interleukin (IL)-15 on purified γδ T cells and its use as a stimulatory signal in the ex vivo expansion of γδ T cells for adoptive transfer. The expansion protocol was validated both with immune cells of healthy individuals and acute myeloid leukemia patients. RESULTS: We report that the addition of IL-15 to γδ T cell cultures results in a more activated phenotype, a higher proliferative capacity, a more pronounced T helper 1 polarization, and an increased cytotoxic capacity of γδ T cells. Moreover γδ T cell expansion starting with peripheral blood mononuclear cells from healthy individuals and acute myeloid leukemia patients is boosted in the presence of IL-15, whereby the antitumor properties of the γδ T cells are strengthened as well. CONCLUSIONS: Our results support the rationale to explore the use of IL-15 in clinical adoptive therapy protocols exploiting γδ T cells.

14.
Oncotarget ; 7(45): 73960-73970, 2016 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-27659531

RESUMEN

We formerly demonstrated that vaccination with Wilms' tumor 1 (WT1)-loaded autologous monocyte-derived dendritic cells (mo-DCs) can be a well-tolerated effective treatment in acute myeloid leukemia (AML) patients. Here, we investigated whether we could introduce the receptor for hyaluronic acid-mediated motility (RHAMM/HMMR/CD168), another clinically relevant tumor-associated antigen, into these mo-DCs through mRNA electroporation and elicit RHAMM-specific immune responses. While RHAMM mRNA electroporation significantly increased RHAMM protein expression by mo-DCs, our data indicate that classical mo-DCs already express and present RHAMM at sufficient levels to activate RHAMM-specific T cells, regardless of electroporation. Moreover, we found that RHAMM-specific T cells are present at vaccination sites in AML patients. Our findings implicate that we and others who are using classical mo-DCs for cancer immunotherapy are already vaccinating against RHAMM.


Asunto(s)
Presentación de Antígeno/inmunología , Antígenos de Neoplasias/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Proteínas de la Matriz Extracelular/inmunología , Receptores de Hialuranos/inmunología , Linfocitos T/inmunología , Vacunas contra el Cáncer/inmunología , Electroporación , Proteínas de la Matriz Extracelular/genética , Expresión Génica , Antígenos HLA-A/inmunología , Humanos , Receptores de Hialuranos/genética , Inmunoterapia , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/terapia , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/terapia , ARN Mensajero/genética , ARN Mensajero/metabolismo , Linfocitos T/metabolismo
15.
Oncotarget ; 6(42): 44123-33, 2015 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-26675759

RESUMEN

In cancer immunotherapy, the use of dendritic cell (DC)-based vaccination strategies can improve overall survival, but until now durable clinical responses remain scarce. To date, DC vaccines are designed primarily to induce effective T-cell responses, ignoring the antitumor activity potential of natural killer (NK) cells. Aiming to further improve current DC vaccination outcome, we engineered monocyte-derived DC to produce interleukin (IL)-15 and/or IL-15 receptor alpha (IL-15Rα) using mRNA electroporation. The addition of IL-15Rα to the protocol, enabling IL-15 transpresentation to neighboring NK cells, resulted in significantly better NK-cell activation compared to IL-15 alone. Next to upregulation of NK-cell membrane activation markers, IL-15 transpresentation resulted in increased NK-cell secretion of IFN-γ, granzyme B and perforin. Moreover, IL-15-transpresenting DC/NK cell cocultures from both healthy donors and acute myeloid leukemia (AML) patients in remission showed markedly enhanced cytotoxic activity against NK cell sensitive and resistant tumor cells. Blocking IL-15 transpresentation abrogated NK cell-mediated cytotoxicity against tumor cells, pointing to a pivotal role of IL-15 transpresentation by IL-15Rα to exert its NK cell-activating effects. In conclusion, we report an attractive approach to improve antitumoral NK-cell activity in DC-based vaccine strategies through the use of IL-15/IL-15Rα mRNA-engineered designer DC.


Asunto(s)
Comunicación Celular , Células Dendríticas/metabolismo , Inmunoterapia Adoptiva/métodos , Interleucina-15/metabolismo , Células Asesinas Naturales/metabolismo , Leucemia Mieloide Aguda/terapia , Activación de Linfocitos , ARN Mensajero/metabolismo , Receptores de Interleucina-15/metabolismo , Vacunas contra el Cáncer/inmunología , Técnicas de Cocultivo , Citotoxicidad Inmunológica , Células Dendríticas/inmunología , Células Dendríticas/trasplante , Electroporación , Ingeniería Genética , Granzimas/metabolismo , Humanos , Interferón gamma/metabolismo , Interleucina-15/genética , Células K562 , Células Asesinas Naturales/inmunología , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/inmunología , Perforina/metabolismo , Fenotipo , ARN Mensajero/genética , Receptores de Interleucina-15/genética , Inducción de Remisión , Transducción de Señal , Factores de Tiempo , Transfección
16.
PLoS One ; 10(5): e0123340, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25951230

RESUMEN

The contribution of natural killer (NK) cells to the treatment efficacy of dendritic cell (DC)-based cancer vaccines is being increasingly recognized. Much current efforts to optimize this form of immunotherapy are therefore geared towards harnessing the NK cell-stimulatory ability of DCs. In this study, we investigated whether generation of human monocyte-derived DCs with interleukin (IL)-15 followed by activation with a Toll-like receptor stimulus endows these DCs, commonly referred to as "IL-15 DCs", with the capacity to stimulate NK cells. In a head-to-head comparison with "IL-4 DCs" used routinely for clinical studies, IL-15 DCs were found to induce a more activated, cytotoxic effector phenotype in NK cells, in particular in the CD56bright NK cell subset. With the exception of GM-CSF, no significant enhancement of cytokine/chemokine secretion was observed following co-culture of NK cells with IL-15 DCs. IL-15 DCs, but not IL-4 DCs, promoted NK cell tumoricidal activity towards both NK-sensitive and NK-resistant targets. This effect was found to require cell-to-cell contact and to be mediated by DC surface-bound IL-15. This study shows that DCs can express a membrane-bound form of IL-15 through which they enhance NK cell cytotoxic function. The observed lack of membrane-bound IL-15 on "gold-standard" IL-4 DCs and their consequent inability to effectively promote NK cell cytotoxicity may have important implications for the future design of DC-based cancer vaccine studies.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Interleucina-15/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Antígeno CD56/metabolismo , Comunicación Celular , Línea Celular Tumoral , Técnicas de Cocultivo , Citocinas/metabolismo , Humanos , Inmunoterapia , Activación de Linfocitos
17.
Cancer Immunol Immunother ; 64(7): 831-42, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25863943

RESUMEN

Dendritic cell (DC) vaccination has demonstrated potential in clinical trials as a new effective cancer treatment, but objective and durable clinical responses are confined to a minority of patients. Interferon (IFN)-α, a type-I IFN, can bolster anti-tumor immunity by restoring or increasing the function of DCs, T cells and natural killer (NK) cells. Moreover, type-I IFN signaling on DCs was found to be essential in mice for tumor rejection by the innate and adaptive immune system. Targeted delivery of IFN-α by DCs to immune cells could boost the generation of anti-tumor immunity, while avoiding the side effects frequently associated with systemic administration. Naturally circulating plasmacytoid DCs, major producers of type-I IFN, were already shown capable of inducing tumor antigen-specific T cell responses in cancer patients without severe toxicity, but their limited number complicates their use in cancer vaccination. In the present work, we hypothesized that engineering easily generated human monocyte-derived mature DCs to secrete IFN-α using mRNA electroporation enhances their ability to promote adaptive and innate anti-tumor immunity. Our results show that IFN-α mRNA electroporation of DCs significantly increases the stimulation of tumor antigen-specific cytotoxic T cell as well as anti-tumor NK cell effector functions in vitro through high levels of IFN-α secretion. Altogether, our findings mark IFN-α mRNA-electroporated DCs as potent inducers of both adaptive and innate anti-tumor immunity and pave the way for clinical trial evaluation in cancer patients.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Interferón-alfa/metabolismo , Proteínas WT1/inmunología , Antígenos de Neoplasias/genética , Linfocitos T CD4-Positivos/inmunología , Proliferación Celular/genética , Células Cultivadas , Células Dendríticas/citología , Células Dendríticas/trasplante , Electroporación , Humanos , Inmunoterapia Adoptiva , Interferón-alfa/genética , Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Neoplasias/inmunología , ARN Mensajero/administración & dosificación , ARN Mensajero/genética , Proteínas WT1/genética
18.
Cytokine Growth Factor Rev ; 26(1): 15-24, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25306466

RESUMEN

Interleukin (IL)-15 is one of the most promising molecules to be used in antitumor immune therapy, as it is able to stimulate the main killer cells of both the innate and adaptive immune system. Although this cytokine can be used as a stand-alone immunotherapeutic agent, IL-15 will probably be most efficient in combination with other strategies to overcome high tumor burden, immune suppression of the tumor microenvironment and/or the short half-life of IL-15. In this review, we will discuss the combination strategies with IL-15 that have been tested to date in different animal tumor models, which include chemotherapy, other immunostimulatory cytokines, targeted therapy, adoptive cell transfer and gene therapy. In addition, we give an overview of IL-15 combination therapies that are currently tested in clinical studies to treat patients with hematological or advanced solid tumors.


Asunto(s)
Citocinas/uso terapéutico , Neoplasias Hematológicas/terapia , Interleucina-15/uso terapéutico , Neoplasias/terapia , Traslado Adoptivo , Animales , Terapia Combinada , Terapia Genética , Semivida , Humanos , Inmunoterapia
19.
J Cell Mol Med ; 18(7): 1372-80, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24979331

RESUMEN

Cervarix™ is approved as a preventive vaccine against infection with the human papillomavirus (HPV) strains 16 and 18, which are causally related to the development of cervical cancer. We are the first to investigate in vitro the effects of this HPV vaccine on interleukin (IL)-15 dendritic cells (DC) as proxy of a naturally occurring subset of blood DC, and natural killer (NK) cells, two innate immune cell types that play an important role in antitumour immunity. Our results show that exposure of IL-15 DC to the HPV vaccine results in increased expression of phenotypic maturation markers, pro-inflammatory cytokine production and cytotoxic activity against HPV-positive tumour cells. These effects are mediated by the vaccine adjuvant, partly through Toll-like receptor 4 activation. Next, we demonstrate that vaccine-exposed IL-15 DC in turn induce phenotypic activation of NK cells, resulting in a synergistic cytotoxic action against HPV-infected tumour cells. Our study thus identifies a novel mode of action of the HPV vaccine in boosting innate immunity, including killing of HPV-infected cells by DC and NK cells.


Asunto(s)
Células Dendríticas/inmunología , Células Asesinas Naturales/inmunología , Papillomaviridae/inmunología , Infecciones por Papillomavirus/inmunología , Vacunas contra Papillomavirus/uso terapéutico , Linfocitos T Citotóxicos/inmunología , Neoplasias del Cuello Uterino/inmunología , Células Cultivadas , Células Dendríticas/metabolismo , Células Dendríticas/patología , Femenino , Humanos , Inmunidad Innata/inmunología , Inmunofenotipificación , Interleucina-15/inmunología , Interleucina-15/metabolismo , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/patología , Linfocitos/inmunología , Linfocitos/metabolismo , Linfocitos/patología , Infecciones por Papillomavirus/patología , Infecciones por Papillomavirus/prevención & control , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/patología , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/prevención & control
20.
Dalton Trans ; 43(25): 9612-9, 2014 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-24831780

RESUMEN

The metal-organic framework MFU-4 shows preferential adsorption of CO2 over N2. This cannot be explained in terms of pore size only. Computational modelling suggests that the unique structure and flexibility of its small 8Cl-cube pore shows a unique gate-diffusion behaviour with different responses to CO2 and N2.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA