Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Pharmaceutics ; 13(3)2021 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-33809779

RESUMEN

Messenger RNA (mRNA) electroporation is a powerful tool for transient genetic modification of cells. This non-viral method of genetic engineering has been widely used in immunotherapy. Electroporation allows fine-tuning of transfection protocols for each cell type as well as introduction of multiple protein-coding mRNAs at once. As a pioneering group in mRNA electroporation, in this review, we provide an expert overview of the ins and outs of mRNA electroporation, discussing the different parameters involved in mRNA electroporation as well as the production of research-grade and production and application of clinical-grade mRNA for gene transfer in the context of cell-based immunotherapies.

2.
Front Immunol ; 10: 1613, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31379824

RESUMEN

Chimeric antigen receptor (CAR)-modified T cell therapy is a rapidly emerging immunotherapeutic approach that is revolutionizing cancer treatment. The impressive clinical results obtained with CAR-T cell therapy in patients with acute lymphoblastic leukemia and lymphoma have fueled the development of CAR-T cells targeting other malignancies, including multiple myeloma (MM). The field of CAR-T cell therapy for MM is still in its infancy, but remains promising. To date, most studies have been performed with B cell maturation antigen (BCMA)-targeted CARs, for which high response rates have been obtained in early-phase clinical trials. However, responses are usually temporary, and relapses have frequently been observed. One of the major reasons for relapse is the loss or downregulation of BCMA expression following CAR-T therapy. This has fostered a search for alternative target antigens that are expressed on the MM cell surface. In this review, we provide an overview of myeloma target antigens other than BCMA that are currently being evaluated in pre-clinical and clinical studies.


Asunto(s)
Antígeno de Maduración de Linfocitos B/inmunología , Mieloma Múltiple/inmunología , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Animales , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Humanos , Inmunoterapia Adoptiva/métodos , Receptores de Antígenos de Linfocitos T/inmunología
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.
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
6.
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
7.
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
8.
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
9.
Transplantation ; 99(1): 120-7, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25050468

RESUMEN

BACKGROUND: Infection with human cytomegalovirus (CMV) is a significant cause of morbidity and mortality in solid organ and hematopoietic stem cell transplant (HSCT) recipients. METHODS: The present study explored the safety, feasibility, and immunogenicity of CMV pp65 messenger RNA-loaded autologous monocyte-derived dendritic cells (DC) as a cellular vaccine for active immunization in healthy volunteers and allogeneic HSCT recipients. Four CMV-seronegative healthy volunteers and three allogeneic HSCT recipients were included in the study. Four clinical-grade autologous monocyte-derived DC vaccines were prepared after a single leukapheresis procedure and administered intradermally at a weekly interval. RESULTS: De novo induction of CMV-specific T-cell responses was detected in three of four healthy volunteers without serious adverse events. Of the HSCT recipients, none developed CMV disease and one of two patients displayed a remarkable threefold increase in CMV pp65-specific T cells on completion of the DC vaccination trial. CONCLUSION: In conclusion, our DC vaccination strategy induced or expanded a CMV-specific cellular response in four of six efficacy-evaluable study subjects, providing a base for its further exploration in larger cohorts.


Asunto(s)
Infecciones por Citomegalovirus/prevención & control , Vacunas contra Citomegalovirus/administración & dosificación , Citomegalovirus/inmunología , Células Dendríticas/trasplante , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Fosfoproteínas/inmunología , ARN Mensajero/biosíntesis , ARN Viral/biosíntesis , Linfocitos T/inmunología , Transfección , Proteínas de la Matriz Viral/inmunología , Adulto , Bélgica , Citomegalovirus/genética , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/virología , Vacunas contra Citomegalovirus/efectos adversos , Vacunas contra Citomegalovirus/genética , Vacunas contra Citomegalovirus/inmunología , Células Dendríticas/inmunología , Células Dendríticas/virología , Estudios de Factibilidad , Femenino , Voluntarios Sanos , Humanos , Esquemas de Inmunización , Inyecciones Intradérmicas , Masculino , Persona de Mediana Edad , Fosfoproteínas/biosíntesis , Fosfoproteínas/genética , ARN Mensajero/genética , ARN Viral/metabolismo , Linfocitos T/virología , Factores de Tiempo , Trasplante Homólogo , Resultado del Tratamiento , Vacunación , Proteínas de la Matriz Viral/biosíntesis , Proteínas de la Matriz Viral/genética , Adulto Joven
10.
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
11.
Cytotherapy ; 16(7): 1024-30, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24856897

RESUMEN

BACKGROUND AIMS: Dendritic cell (DC)-based immunotherapy has shown potential to counteract autoimmunity in multiple sclerosis (MS). METHODS: We compared the phenotype and T-cell stimulatory capacity of in vitro generated monocyte-derived DC from MS patients with those from healthy controls. RESULTS: Except for an increase in the number of C-C chemokine receptor 7-expressing DC from MS patients, no major differences were found between groups in the expression of maturation-associated membrane markers or in the in vitro capacity to stimulate autologous T cells. CONCLUSIONS: Our observations may pave the way for the development of patient-tailored DC-based vaccination strategies to treat MS.


Asunto(s)
Células Dendríticas/inmunología , Inmunoterapia , Activación de Linfocitos/inmunología , Esclerosis Múltiple/prevención & control , Receptores CCR7/biosíntesis , Adulto , Anciano , Diferenciación Celular/genética , Células Dendríticas/citología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Monocitos/citología , Monocitos/metabolismo , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Receptores CCR7/inmunología , Linfocitos T/inmunología , Vacunación
12.
Methods Mol Biol ; 1139: 233-41, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24619684

RESUMEN

In this chapter, we describe the technique of electroporation as an efficient method to load primary leukemic cells with the double-stranded RNA (dsRNA) analogue, polyriboinosinic polyribocytidylic acid (poly(I:C)), and detail on the delicate freezing and thawing procedure of primary leukemic cells.Electroporation is a non-viral gene transfer method by which short-term pores in the membrane of cells are generated by an electrical pulse, allowing molecules to enter the cell. RNA electroporation, a technique developed in our laboratory, is a widely used and versatile transfection method for efficient introduction of both coding RNA (messenger RNA) and non-coding RNA, e.g., dsRNA and small interfering (siRNA), into mammalian cells. Accurate cell processing and storage of patient material is essential for optimal recovery and quality of the cell product for downstream applications.


Asunto(s)
Electroporación/métodos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Poli I-C/metabolismo , Criopreservación , Humanos
13.
Mult Scler ; 20(5): 548-57, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24057429

RESUMEN

BACKGROUND: The role of the adaptive immune system and more specifically T cells in the pathogenesis of multiple sclerosis (MS) has been studied extensively. Emerging evidence suggests that dendritic cells (DCs), which are innate immune cells, also contribute to MS. OBJECTIVES: This study aimed to characterize circulating DC populations in MS and to investigate the contribution of MS-associated genetic risk factors to DCs. METHODS: Ex vivo analysis of conventional (cDCs) and plasmacytoid DCs (pDCs) was carried out on peripheral blood of MS patients (n = 110) and age- and gender-matched healthy controls (n = 112). RESULTS: Circulating pDCs were significantly decreased in patients with chronic progressive MS compared to relapsing-remitting MS and healthy controls. While no differences in cDCs frequency were found between the different study groups, HLA-DRB1*1501(+) MS patients and patients not carrying the protective IL-7Rα haplotype 2 have reduced frequencies of circulating cDCs and pDCs, respectively. MS-derived DCs showed enhanced IL-12p70 production upon TLR ligation and had an increased expression of the migratory molecules CCR5 and CCR7 as well as an enhanced in vitro chemotaxis. CONCLUSION: DCs in MS are in a pro-inflammatory state, have a migratory phenotype and are affected by genetic risk factors, thereby contributing to pathogenic responses.


Asunto(s)
Células Dendríticas/inmunología , Inmunidad Innata , Inflamación/genética , Inflamación/inmunología , Esclerosis Múltiple Crónica Progresiva/genética , Esclerosis Múltiple Crónica Progresiva/inmunología , Esclerosis Múltiple Recurrente-Remitente/genética , Esclerosis Múltiple Recurrente-Remitente/inmunología , Adulto , Anciano , Estudios de Casos y Controles , Células Cultivadas , Quimiotaxis , Células Dendríticas/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Cadenas HLA-DRB1/genética , Haplotipos , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Receptores CCR5/metabolismo , Receptores CCR7/metabolismo , Receptores de Interleucina-17/genética , Factores de Riesgo , Receptores Toll-Like/metabolismo , Adulto Joven
14.
Oncologist ; 17(10): 1256-70, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22907975

RESUMEN

The cytotoxic and regulatory antitumor functions of natural killer (NK) cells have become attractive targets for immunotherapy. Manipulation of specific NK cell functions and their reciprocal interactions with dendritic cells (DCs) might hold therapeutic promise. In this review, we focus on the engagement of NK cells in DC-based cancer vaccination strategies, providing a comprehensive overview of current in vivo experimental and clinical DC vaccination studies encompassing the monitoring of NK cells. From these studies, it is clear that NK cells play a key regulatory role in the generation of DC-induced antitumor immunity, favoring the concept that targeting both innate and adaptive immune mechanisms may synergistically promote clinical outcome. However, to date, DC vaccination trials are only infrequently accompanied by NK cell monitoring. Here, we discuss different strategies to improve DC vaccine preparations via exploitation of NK cells and provide a summary of relevant NK cell parameters for immune monitoring. We underscore that the design of DC-based cancer vaccines should include the evaluation of their NK cell stimulating potency both in the preclinical phase and in clinical trials.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Inmunoterapia Adoptiva/métodos , Células Asesinas Naturales/inmunología , Animales , Citotoxicidad Inmunológica , Humanos
15.
Clin Dev Immunol ; 2012: 184979, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22844321

RESUMEN

A variety of immune-based therapies has been developed in order to boost or induce protective CD8(+) T cell responses in order to control HIV replication. Since dendritic cells (DCs) are professional antigen-presenting cells (APCs) with the unique capability to stimulate naïve T cells into effector T cells, their use for the induction of HIV-specific immune responses has been studied intensively. In the present study we investigated whether modulation of the activation state of DCs electroporated with consensus codon-optimized HxB2 gag mRNA enhances their capacity to induce HIV gag-specific T cell responses. To this end, mature DCs were (i) co-electroporated with mRNA encoding interleukin (IL)-12p70 mRNA, or (ii) activated with a cytokine cocktail consisting of R848 and interferon (IFN)-γ. Our results confirm the ability of HxB2 gag-expressing DCs to expand functional HIV-specific CD8(+) T cells. However, although most of the patients had detectable gag-specific CD8(+) T cell responses, no significant differences in the level of expansion of functional CD8(+) T cells could be demonstrated when comparing conventional or immune-modulated DCs expressing IL-12p70. This result which goes against expectation may lead to a re-evaluation of the need for IL-12 expression by DCs in order to improve T-cell responses in HIV-1-infected individuals.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Infecciones por VIH/inmunología , VIH-1 , Interleucina-12/metabolismo , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/inmunología , Presentación de Antígeno , Células Dendríticas/metabolismo , Electroporación , Técnicas de Transferencia de Gen , Infecciones por VIH/metabolismo , Infecciones por VIH/terapia , VIH-1/inmunología , Humanos , Imidazoles/farmacología , Interferón gamma/farmacología , Interleucina-12/genética , Activación de Linfocitos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética
16.
Oncologist ; 17(2): 250-9, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22291091

RESUMEN

There is a growing body of evidence that Wilms' tumor protein 1 (WT1) is a promising tumor antigen for the development of a novel class of universal cancer vaccines. Recently, in a National Cancer Institute prioritization project, WT1 was ranked first in a list of 75 cancer antigens. In this light, we exhaustively reviewed all published cancer vaccine trials reporting on WT1-targeted active specific immunotherapy in patients with hematological malignancies and solid tumors. In all clinical trials, vaccine-induced immunological responses could be detected. Importantly, objective clinical responses (including stable disease) were observed in 46% and 64% of evaluable vaccinated patients with solid tumors and hematological malignancies, respectively. Immunogenicity of WT1-based cancer vaccines was demonstrated by the detection of a specific immunological response in 35% and 68% of evaluable patients with solid tumors and hematological malignancies, respectively. In order to become part of the armamentarium of the modern oncologist, it will be important to design WT1-based immunotherapies applicable to a large patient population, to standardize vaccination protocols enabling systematic review, and to further optimize the immunostimulatory capacity of the vaccine components. Moreover, improved immunomonitoring tools that reveal clinically relevant T-cell responses will further shape the ideal WT1 immunotherapy strategy. In conclusion, the clinical results obtained so far in WT1-targeted cancer vaccine trials reveal an untapped potential for inducing cancer immunity with minimal side effects and hold promise for a new adjuvant treatment against residual disease and against cancer relapse.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Neoplasias Hematológicas/terapia , Inmunoterapia Activa , Neoplasias/terapia , Proteínas WT1/inmunología , Ensayos Clínicos como Asunto , Neoplasias Hematológicas/inmunología , Humanos , Inmunoterapia Activa/efectos adversos
17.
AIDS ; 26(4): F1-12, 2012 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-22156965

RESUMEN

BACKGROUND: In an effort to raise protective antiviral immunity, dendritic cell immunotherapy was evaluated in six adults infected with human immunodeficiency virus (HIV)-1 and stable under highly active antiretroviral therapy (HAART). DESIGN AND METHODS: Autologous monocyte-derived dendritic cells electroporated with mRNA encoding Gag and a chimeric Tat-Rev-Nef protein were administered, whereas patients remained on HAART. Feasibility, safety, immunogenicity and antiviral responses were investigated. RESULTS: Dendritic cell vaccine preparation and administration were successful in all patients and only mild adverse events were seen. There was a significant increase post-dendritic cell as compared to pre-dendritic cell vaccination in magnitude and breadth of HIV-1-specific interferon (IFN)-γ response, in particular to Gag, and in T-cell proliferation. Breadth of IFN-γ response and T-cell proliferation were both correlated with CD4(+) and CD8(+) polyfunctional T-cell responses. Importantly, dendritic cell vaccination induced or increased the capacity of autologous CD8(+) T cells to inhibit superinfection of CD4(+) T cells with the vaccine-related IIIB virus and some but not all other HIV-1 strains tested. This HIV-1-inhibitory activity, indicative of improved antiviral response, was correlated with magnitude and breadth of Gag-specific IFN-γ response. CONCLUSIONS: Therapeutic immunization with dendritic cells was safe and successful in raising antiviral cellular immune responses, including effector CD8(+) T cells with virus inhibitory activity. The stimulation of those potent immunological and antiviral effects, which have been associated with control of HIV-1, underscores the potential of dendritic cell vaccination in the treatment of HIV-1. The incomplete nature of the response in some patients helped to identify potential targets for future improvement, that is increasing antigenic spectrum and enhancing T-cell response.


Asunto(s)
Vacunas contra el SIDA/inmunología , Antivirales/inmunología , Células Dendríticas/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Proteínas del Virus de la Inmunodeficiencia Humana/inmunología , Activación de Linfocitos , ARN Mensajero/inmunología , Linfocitos T/inmunología , Vacunas contra el SIDA/administración & dosificación , Vacunas contra el SIDA/efectos adversos , Adulto , Terapia Antirretroviral Altamente Activa , Antivirales/administración & dosificación , Antivirales/efectos adversos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Electroporación , Estudios de Factibilidad , Humanos , Interferón gamma/inmunología , Activación de Linfocitos/inmunología , Masculino , Persona de Mediana Edad , ARN Viral/inmunología , Proteínas Recombinantes de Fusión/inmunología , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/inmunología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/inmunología , Productos del Gen rev del Virus de la Inmunodeficiencia Humana/inmunología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/inmunología
18.
PLoS One ; 6(6): e20952, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21698118

RESUMEN

α active specific immunotherapy aims at stimulating the host's immune system to recognize and eradicate malignant cells. The concomitant activation of dendritic cells (DC) and natural killer (NK) cells is an attractive modality for immune-based therapies. Inducing immunogenic cell death to facilitate tumor cell recognition and phagocytosis by neighbouring immune cells is of utmost importance for guiding the outcome of the immune response. We previously reported that acute myeloid leukemic (AML) cells in response to electroporation with the synthetic dsRNA analogue poly(I:C) exert improved immunogenicity, demonstrated by enhanced DC-activating and NK cell interferon-γ-inducing capacities. To further invigorate the potential of these immunogenic tumor cells, we explored their effect on the phagocytic and cytotoxic capacity of DC and NK cells, respectively. Using single-cell analysis, we assessed these functionalities in two- and three-party cocultures. Following poly(I:C) electroporation AML cells become highly susceptible to NK cell-mediated killing and phagocytosis by DC. Moreover, the enhanced killing and the improved uptake are strongly correlated. Interestingly, tumor cell killing, but not phagocytosis, is further enhanced in three-party cocultures provided that these tumor cells were upfront electroporated with poly(I:C). Altogether, poly(I:C)-electroporated AML cells potently activate DC and NK cell functions and stimulate NK-DC cross-talk in terms of tumor cell killing. These data strongly support the use of poly(I:C) as a cancer vaccine component, providing a way to overcome immune evasion by leukemic cells.


Asunto(s)
Leucemia Mieloide Aguda/patología , Poli I-C/farmacología , Células Dendríticas , Electroporación , Colorantes Fluorescentes , Humanos , Células Asesinas Naturales , Leucemia Mieloide Aguda/inmunología , Fagocitosis
19.
Cancer Immunol Immunother ; 60(6): 757-69, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21519825

RESUMEN

Immunotherapy is currently under active investigation as an adjuvant therapy to improve the overall survival of patients with acute myeloid leukaemia (AML) by eliminating residual leukaemic cells following standard therapy. The graft-versus-leukaemia effect observed following allogeneic haematopoietic stem cell transplantation has already demonstrated the significant role of immune cells in controlling AML, paving the way to further exploitation of this effect in optimized immunotherapy protocols. In this review, we discuss the current state of cellular immunotherapy as adjuvant therapy for AML, with a particular focus on new strategies and recently published results of preclinical and clinical studies. Therapeutic vaccines that are being tested in AML include whole tumour cells as an autologous source of multiple leukaemia-associated antigens (LAA) and autologous dendritic cells loaded with LAA as effective antigen-presenting cells. Furthermore, adoptive transfer of cytotoxic T cells or natural killer cells is under active investigation. Results from phase I and II trials are promising and support further investigation into the potential of cellular immunotherapeutic strategies to prevent or fight relapse in AML patients.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Leucemia Mieloide Aguda/terapia , Humanos , Leucemia Mieloide Aguda/inmunología
20.
Eur J Hum Genet ; 19(9): 965-73, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21522185

RESUMEN

DFNA5 was first identified as a gene causing autosomal dominant hearing loss (HL). Different mutations have been found, all exerting a highly specific gain-of-function effect, in which skipping of exon 8 causes the HL. Later reports revealed the involvement of the gene in different types of cancer. Epigenetic silencing of DFNA5 in a large percentage of gastric, colorectal and breast tumors and p53-dependent transcriptional activity have been reported, concluding that DFNA5 acts as a tumor suppressor gene in different frequent types of cancer. Despite these data, the molecular function of DFNA5 has not been investigated properly. Previous transfection studies with mutant DFNA5 in yeast and in mammalian cells showed a toxic effect of the mutant protein, which was not seen after transfection of the wild-type protein. Here, we demonstrate that DFNA5 is composed of two domains, separated by a hinge region. The first region induces apoptosis when transfected in HEK293T cells, the second region masks and probably regulates this apoptosis inducing capability. Moreover, the involvement of DFNA5 in apoptosis-related pathways in a physiological setting was demonstrated through gene expression microarray analysis using Dfna5 knockout mice. In view of its important role in carcinogenesis, this finding is expected to lead to new insights on the role of apoptosis in many types of cancer. In addition, it provides a new line of evidence supporting an important role for apoptosis in monogenic and complex forms of HL.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Genes Supresores de Tumor/fisiología , Pérdida Auditiva/genética , Neoplasias/genética , Receptores de Estrógenos/genética , Animales , Apoptosis/genética , Sordera/genética , Exones , Células HEK293 , Células HeLa , Humanos , Ratones , Ratones Noqueados , Estructura Terciaria de Proteína , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA