Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Ther Oncolytics ; 24: 43-58, 2022 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-34977341

RESUMEN

PD-1/PD-L1 checkpoint blockade has achieved unprecedented success in cancer immunotherapy. Nevertheless, many immune-excluded tumors are resistant to therapy. Combination with oncolytic virotherapy may overcome resistance by inducing acute inflammation, immune cell recruitment, and remodeling of the tumor immune environment. Here, we assessed the combination of oncolytic measles vaccine (MV) vectors and PD-1/PD-L1 blockade. In the MC38cea model of measles virus oncolysis, MV combined with anti-PD-1 and MV vectors encoding anti-PD-1 or anti-PD-L1 antibodies achieved modest survival benefits compared with control MV or vectors encoding the antibody constant regions only. Analyses of tumor samples and tumor-draining lymph nodes revealed slight increases in intratumoral T cell effector cytokines as well as a shift toward an effector memory phenotype in the T cell compartment. Importantly, increased IFN-γ recall responses were observed in tumor rechallenge experiments with mice in complete tumor remission after treatment with MV encoding anti-PD-1 or anti-PD-L1 compared with control MV. These results prompted us to generate MV encoding the clinically approved agents pembrolizumab and nivolumab. Previously, we have generated MV encoding atezolizumab. We demonstrated the functionality of the novel vectors in vitro. We envision these vectors as therapeutics that induce and support durable anti-tumor immune memory.

2.
Front Immunol ; 13: 1096162, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36726983

RESUMEN

Introduction: Pancreatic ductal adenocarcinoma (PDAC) is largely refractory to cancer immunotherapy with PD-1 immune checkpoint blockade (ICB). Oncolytic virotherapy has been shown to synergize with ICB. In this work, we investigated the combination of anti-PD-1 and oncolytic measles vaccine in an immunocompetent transplantable PDAC mouse model. Methods: We characterized tumor-infiltrating T cells by immunohistochemistry, flow cytometry and T cell receptor sequencing. Further, we performed gene expression profiling of tumor samples at baseline, after treatment, and when tumors progressed. Moreover, we analyzed systemic anti-tumor and anti-viral immunity. Results: Combination treatment significantly prolonged survival compared to monotherapies. Tumor-infiltrating immune cells were increased after virotherapy. Gene expression profiling revealed a unique, but transient signature of immune activation after combination treatment. However, systemic anti-tumor immunity was induced by virotherapy and remained detectable even when tumors progressed. Anti-PD-1 treatment did not impact anti-viral immunity. Discussion: Our results indicate that combined virotherapy and ICB induces anti-tumor immunity and reshapes the tumor immune environment. However, further refinement of this approach may be required to develop its full potential and achieve durable efficacy.


Asunto(s)
Carcinoma Ductal Pancreático , Viroterapia Oncolítica , Neoplasias Pancreáticas , Ratones , Animales , Neoplasias Pancreáticas/patología , Carcinoma Ductal Pancreático/genética , Inmunoterapia/métodos , Viroterapia Oncolítica/métodos , Neoplasias Pancreáticas
3.
Methods Mol Biol ; 2058: 213-227, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31486040

RESUMEN

Oncolytic virotherapy is rapidly advancing into clinical practice as an appealing strategy for cancer therapy. Studies in the recent decades indicate that immunostimulatory properties of oncolytic viruses (OVs) are crucial for their therapeutic efficacy. The specific lysis of tumor cells and release of tumor associated antigens in the context of an OV infection ensures activation of a tumor-specific immune response. The evidence for induction of a systemic, specific antitumor immune response after OV therapy in preclinical and clinical studies allows to consider oncolytic virotherapy as a type of therapeutic cancer vaccination strategy.This chapter describes methods to evaluate the therapeutic tumor vaccination effect of an oncolytic virus in murine tumor models. Protocols for a tumor rechallenge experiment in vivo and tumor cell specific splenocyte restimulation in vitro are included, as well as protocols for analysis of memory T cell subpopulations in tumor draining lymph nodes using flow cytometry.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Vectores Genéticos/genética , Neoplasias/inmunología , Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/genética , Animales , Vacunas contra el Cáncer/genética , Línea Celular Tumoral , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Memoria Inmunológica , Inmunofenotipificación , Activación de Linfocitos/inmunología , Ratones , Neoplasias/metabolismo , Neoplasias/patología , Bazo/inmunología , Bazo/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Vacunación , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Viruses ; 11(10)2019 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-31623390

RESUMEN

Tumor-targeted immunomodulation using oncolytic viral vectors is currently being investigated as a promising strategy in cancer therapy. In a previous study, we showed that a measles virus Schwarz vaccine strain (MeVac) vector encoding an interleukin-12 fusion protein (FmIL-12) is an effective immunotherapy in the MC38cea murine colon adenocarcinoma model. We hypothesized that MeVac encoding interleukin-15 may mediate enhanced T and NK cell responses and thus increase the therapeutic efficacy, especially in NK cell-controlled tumors. Therefore, we generated MeVac vectors encoding an interleukin-15 superagonist, FmIL-15. Replication and oncolytic capacity, transgene expression, and functionality of MeVac FmIL-15 vectors were validated in vitro. Effects on the tumor immune landscape and therapeutic efficacy of both FmIL-12 and FmIL-15 vectors were studied in the MC38cea and B16hCD46 tumor models. Treatment with MeVac FmIL-15 increased T and NK cell infiltration in both models. However, MeVac FmIL-12 showed more robust viral gene expression and immune activation, resulting in superior anti-tumor efficacy. Based on these results, MeVac encoding a human IL-12 fusion protein was developed for future clinical translation.


Asunto(s)
Regulación Viral de la Expresión Génica , Interleucina-12/agonistas , Interleucina-15/agonistas , Vacuna Antisarampión/inmunología , Adenocarcinoma , Animales , Línea Celular Tumoral , Supervivencia Celular , Colon , Modelos Animales de Enfermedad , Femenino , Genes Virales , Inmunoterapia , Interleucina-12/genética , Interleucina-15/genética , Células Asesinas Naturales/inmunología , Sarampión , Ratones , Ratones Endogámicos C57BL , Virus Oncolíticos , Transcriptoma , Vacunas Sintéticas , Proteínas Virales de Fusión/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Clin Cancer Res ; 24(9): 2128-2137, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29437789

RESUMEN

Purpose: Immunotherapy with bispecific T-cell engagers has achieved striking success against hematologic malignancies, but efficacy against solid tumors has been limited. We hypothesized that oncolytic measles viruses encoding bispecific T-cell engagers (MV-BiTEs) represent a safe and effective treatment against solid tumors through local BiTE expression, direct tumor cell lysis and in situ tumor vaccination.Experimental Design: To test this hypothesis, we generated MV-BiTEs from the Edmonston B vaccine strain to target two model antigens. Replicative and oncolytic potential were assessed by infection and cell viability assays, respectively. Functionality of virus-derived BiTEs was tested in vitro by complementary binding and cytotoxicity assays. In vivo efficacy of MV-BiTE was investigated using both syngeneic and xenograft mouse models of solid cancers.Results: We verified secretion of functional BiTE antibodies by MV-BiTE-infected cells. Further, we demonstrated therapeutic efficacy of MV-BiTE against established tumors in fully immunocompetent mice. MV-BiTE efficacy was associated with increased intratumoral T-cell infiltration and induction of protective antitumor immunity. In addition, we showed therapeutic efficacy of MV-BiTE in xenograft models of patient-derived primary colorectal carcinoma spheroids with transfer of peripheral blood mononuclear cells.Conclusions: MV-BiTE treatment was effective in two distinct models of solid tumors without signs of toxicity. This provides strong evidence for therapeutic benefits of tumor-targeted BiTE expression by oncolytic MV. Thus, this study represents proof of concept for an effective strategy to treat solid tumors with BiTEs. Clin Cancer Res; 24(9); 2128-37. ©2018 AACR.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Expresión Génica , Neoplasias/inmunología , Viroterapia Oncolítica , Virus Oncolíticos/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Vacunas contra el Cáncer/genética , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Inmunoterapia , Ratones , Neoplasias/genética , Neoplasias/patología , Neoplasias/terapia , Virus Oncolíticos/genética , Esferoides Celulares , Especificidad del Receptor de Antígeno de Linfocitos T , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Gen Virol ; 98(9): 2248-2257, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28809150

RESUMEN

Viruses from the diverse family of Paramyxoviridae include important pathogens and are applied in gene therapy and for cancer treatment. The Tupaia paramyxovirus (TPMV), isolated from the kidney of a tree shrew, does not infect human cells and neutralizing antibodies against other Paramyxoviridae do not cross-react with TPMV. Here, we present a vector system for de novo generation of infectious TPMV that allows for insertion of additional genes as well as targeting using antibody single-chain variable fragments. We show that the recombinant TPMV specifically infect cells expressing the targeted receptor and replicate in human cells. This vector system provides a valuable tool for both basic research and therapeutic applications.


Asunto(s)
Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Paramyxoviridae/genética , Animales , Línea Celular , Vectores Genéticos/fisiología , Humanos , Paramyxoviridae/fisiología , Transgenes , Tupaia/virología
7.
Oncoimmunology ; 6(4): e1285992, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28507792

RESUMEN

Combination of oncolytic virotherapy with immunomodulators is emerging as a promising therapeutic strategy for numerous tumor entities. In this study, we developed measles Schwarz vaccine strain vectors encoding immunomodulators to support different phases in the establishment of antitumor immune responses. Therapeutic efficacy of the novel vectors was evaluated in the immunocompetent MC38cea tumor model. We identified vectors encoding an IL-12 fusion protein (MeVac FmIL-12) and an antibody against PD-L1 (MeVac anti-PD-L1), respectively, as the most effective. Treatment of established tumors with MeVac FmIL-12 achieved 90% complete remissions. Profiling of the tumor immune microenvironment revealed activation of a type 1 T helper cell-directed response, with MeVac FmIL-12 ensuring potent early natural killer and effector T cell activation as well as upregulation of the effector cytokines IFN-γ and TNF-α. CD8+ T cells were found to be essential for the therapeutic efficacy of MeVac FmIL-12. Results of this study present MeVac FmIL-12 as a novel approach for targeted IL-12 delivery and elucidate mechanisms of successful immunovirotherapy.

8.
Mol Ther ; 22(11): 1949-59, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25156126

RESUMEN

We hypothesized that the combination of oncolytic virotherapy with immune checkpoint modulators would reduce tumor burden by direct cell lysis and stimulate antitumor immunity. In this study, we have generated attenuated Measles virus (MV) vectors encoding antibodies against CTLA-4 and PD-L1 (MV-aCTLA-4 and MV-aPD-L1). We characterized the vectors in terms of growth kinetics, antibody expression, and cytotoxicity in vitro. Immunotherapeutic effects were assessed in a newly established, fully immunocompetent murine model of malignant melanoma, B16-CD20. Analyses of tumor-infiltrating lymphocytes and restimulation experiments indicated a favorable immune profile after MV-mediated checkpoint modulation. Therapeutic benefits in terms of delayed tumor progression and prolonged median overall survival were observed for animals treated with vectors encoding anti-CTLA-4 and anti-PD-L1, respectively. Combining systemic administration of antibodies with MV treatment also improved therapeutic outcome. In vivo oncolytic efficacy against human tumors was studied in melanoma xenografts. MV-aCTLA-4 and MV-aPD-L1 were equally efficient as parental MV in this model, with high rates of complete tumor remission (> 80%). Furthermore, we could demonstrate lysis of tumor cells and transgene expression in primary tissue from melanoma patients. The current results suggest rapid translation of combining immune checkpoint modulation with oncolytic viruses into clinical application.


Asunto(s)
Antígeno B7-H1/metabolismo , Antígeno CTLA-4/metabolismo , Linfocitos Infiltrantes de Tumor/metabolismo , Melanoma Experimental/terapia , Virus Oncolíticos/inmunología , Animales , Vectores Genéticos/administración & dosificación , Virus del Sarampión/genética , Virus del Sarampión/inmunología , Virus del Sarampión/metabolismo , Melanoma Experimental/inmunología , Ratones , Viroterapia Oncolítica , Virus Oncolíticos/genética , Virus Oncolíticos/metabolismo , Resultado del Tratamiento
9.
Biotechnol Appl Biochem ; 61(1): 65-73, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-23941496

RESUMEN

Double-stranded RNA (dsRNA) is a pathogen-associated molecular pattern, known for its ability to induce antiviral response and enhance communication between cells mediating innate and adaptive immune responses. The aim of this study was to characterize the effect of the dsRNA-containing product Larifan on the production of a wide spectrum of cytokines and chemokines in ex vivo cultivated peripheral blood mononuclear cells. Concentrations of 29 different cytokines were detected by a Luminex® 200™ System using three Milliplex MAP Multiplex Assay Kits. Larifan caused strong induction of chemokine macrophage inflammatory protein 1ß, I-309, and TARC, proinflammatory cytokines IL-6, tumor necrosis factor -α, granulocyte macrophage colony-stimulating factor, anti-inflammatory IL-10, and cellular immunity mediating factors IL-23 and interferon-γ. Considerable suppression of IL-16 and chemokine stromal cell-derived factor 1 a+b and interferon gamma-induced protein 10 was also observed. The network of molecules responding to the presence of Larifan revealed the pleiotropic effect this product exerts on immune response.


Asunto(s)
Citocinas/biosíntesis , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , ARN Bicatenario/farmacología , Adulto , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Compuestos Orgánicos/farmacología
10.
Melanoma Res ; 23(3): 221-6, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23546221

RESUMEN

Hereditary cutaneous melanoma is associated with mutations in the high-risk CDKN2A gene in about 40% of melanoma-prone families. Mutations in the CDK4 gene are the cause in only a few pedigrees. In this study, we analyzed 20 Latvian familial melanoma probands and carried out a comprehensive analysis of CDKN2A including sequencing of its promoter/intronic regions and deletion screening. We also analyzed the critical second exon of the CDK4 gene. One novel intronic variant (IVS2+82C>T) of the CDKN2A gene and a small deletion (c.-20677_-20682delGTACGC) in its promoter region were found. Genotyping of the novel variants in larger melanoma and control groups indicated that the deletion increases the risk of melanoma (odds ratio=6.353, 95% confidence interval: 1.34-30.22, P=0.0168). The CDK4 gene analysis showed a Latvian melanoma family with the mutation R24H carried on the same haplotype as in two previously described Latvian CDK4-positive families. Our study suggests that the main risk gene in Latvian families with a strong family history of melanoma is CDK4 and that most of the other cases analyzed could be sporadic or associated with low-penetrance risk genes.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/genética , Genes p16 , Mutación de Línea Germinal , Melanoma/genética , Neoplasias Cutáneas/genética , Anciano , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Femenino , Efecto Fundador , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Letonia , Masculino , Melanoma/enzimología , Persona de Mediana Edad , Neoplasias Cutáneas/enzimología
11.
J Med Genet ; 50(4): 264-70, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23384855

RESUMEN

BACKGROUND: CDKN2A and CDK4 are high risk susceptibility genes for cutaneous malignant melanoma. Melanoma families with CDKN2A germline mutations have been extensively characterised, whereas CDK4 families are rare and lack a systematic investigation of their phenotype. METHODS: All known families with CDK4 germline mutations (n=17) were recruited for the study by contacting the authors of published papers or by requests via the Melanoma Genetics Consortium (GenoMEL). Phenotypic data related to primary melanoma and pigmentation characteristics were collected. The CDK4 exon 2 and the complete coding region of the MC1R gene were sequenced. RESULTS: Eleven families carried the CDK4 R24H mutation whereas six families had the R24C mutation. The total number of subjects with verified melanoma was 103, with a median age at first melanoma diagnosis of 39 years. Forty-three (41.7%) subjects had developed multiple primary melanomas (MPM). A CDK4 mutation was found in 89 (including 62 melanoma cases) of 209 tested subjects. CDK4 positive family members (both melanoma cases and unaffected subjects) were more likely to have clinically atypical nevi than CDK4 negative family members (p<0.001). MPM subjects had a higher frequency of MC1R red hair colour variants compared with subjects with one tumour (p=0.010). CONCLUSION: Our study shows that families with CDK4 germline mutations cannot be distinguished phenotypically from CDKN2A melanoma families, which are characterised by early onset of disease, increased occurrence of clinically atypical nevi, and development of MPM. In a clinical setting, the CDK4 gene should therefore always be examined when a melanoma family tests negative for CDKN2A mutation.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Color del Cabello/genética , Melanoma/genética , Neoplasias Cutáneas/genética , Adulto , Exones , Femenino , Mutación de Línea Germinal , Humanos , Masculino , Melanoma/patología , Persona de Mediana Edad , Fenotipo , Neoplasias Cutáneas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...