Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Blood Adv ; 3(5): 797-812, 2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30850386

RESUMEN

The oncolytic reovirus (RV) has demonstrated clinical efficacy and minimal toxicity in a variety of cancers, including multiple myeloma (MM). MM is a malignancy of plasma cells that is considered treatable but incurable because of the 90% relapse rate that is primarily from drug resistance. The systemic nature of MM and the antitumor immunosuppression by its tumor microenvironment presents an ongoing therapeutic challenge. In the present study, we demonstrate that RV synergizes with the standard-of-care MM drug bortezomib (BTZ) and, importantly, enhances its therapeutic potential in therapy-resistant human MM cell lines in vitro. Using the syngeneic Vk*MYC BTZ-resistant immunocompetent transplantable MM murine model, we also demonstrate that mice harboring BTZ-insensitive MM tumors respond to the RV/BTZ combination treatment in terms of decreased tumor burden and improved overall survival (P < .00001). We demonstrate that BTZ augments RV replication in tumor-associated endothelial cells and myeloma cells, leading to enhanced viral delivery and thereby stimulating cytokine release, immune activity, apoptosis, and reduction of the MM-associated immune suppression. We conclude that combined RV/BTZ is an attractive therapeutic strategy with no safety signals for the treatment of MM.


Asunto(s)
Bortezomib/uso terapéutico , Terapia Combinada/métodos , Inmunoterapia/métodos , Mieloma Múltiple/terapia , Viroterapia Oncolítica/métodos , Animales , Bortezomib/farmacología , Línea Celular Tumoral , Células Endoteliales/virología , Humanos , Ratones , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/mortalidad , Virus Oncolíticos/inmunología , Terapia Recuperativa/métodos , Replicación Viral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Cancers (Basel) ; 10(6)2018 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-29914097

RESUMEN

As the current efficacy of oncolytic viruses (OVs) as monotherapy is limited, exploration of OVs as part of a broader immunotherapeutic treatment strategy for cancer is necessary. Here, we investigated the ability for immune checkpoint blockade to enhance the efficacy of oncolytic reovirus (RV) for the treatment of breast cancer (BrCa). In vitro, oncolysis and cytokine production were assessed in human and murine BrCa cell lines following RV exposure. Furthermore, RV-induced upregulation of tumor cell PD-L1 was evaluated. In vivo, the immunocompetent, syngeneic EMT6 murine model of BrCa was employed to determine therapeutic and tumor-specific immune responses following treatment with RV, anti-PD-1 antibodies or in combination. RV-mediated oncolysis and cytokine production were observed following BrCa cell infection and RV upregulated tumor cell expression of PD-L1. In vivo, RV monotherapy significantly reduced disease burden and enhanced survival in treated mice, and was further enhanced by PD-1 blockade. RV therapy increased the number of intratumoral regulatory T cells, which was reversed by the addition of PD-1 blockade. Finally, dual treatment led to the generation of a systemic adaptive anti-tumor immune response evidenced by an increase in tumor-specific IFN-γ producing CD8⁺ T cells, and immunity from tumor re-challenge. The combination of PD-1 blockade and RV appears to be an efficacious immunotherapeutic strategy for the treatment of BrCa, and warrants further investigation in early-phase clinical trials.

3.
Clin Cancer Res ; 22(23): 5839-5850, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27220962

RESUMEN

PURPOSE: In addition to their direct cytopathic effects, oncolytic viruses are capable of priming antitumor immune responses. However, strategies to enhance the immunotherapeutic potential of these agents are lacking. Here, we investigated the ability of the multi-tyrosine kinase inhibitor and first-line metastatic renal cell carcinoma (RCC) agent, sunitinib, to augment the antitumor immune response generated by oncolytic reovirus. EXPERIMENTAL DESIGN: In vitro, oncolysis and chemokine production were assessed in a panel of human and murine RCC cell lines after exposure to reovirus, sunitinib, or their combination. In vivo, the RENCA syngeneic murine model of RCC was employed to determine therapeutic and tumor-specific immune responses after treatment with reovirus (intratumoral), sunitinib, or their combination. Parallel investigations employing the KLN205 syngeneic murine model of lung squamous cell carcinoma (NSCLC) were conducted for further validation. RESULTS: Reovirus-mediated oncolysis and chemokine production was observed following RCC infection. Reovirus monotherapy reduced tumor burden and was capable of generating a systemic adaptive antitumor immune response evidenced by increased numbers of tumor-specific CD8+ IFNγ-producing cells. Coadministration of sunitinib with reovirus further reduced tumor burden resulting in improved survival, decreased accumulation of immune suppressor cells, and the establishment of protective immunity upon tumor rechallenge. Similar results were observed for KLN205 tumor-bearing mice, highlighting the potential broad applicability of this approach. CONCLUSIONS: The ability to repurpose sunitinib for augmentation of reovirus' immunotherapeutic efficacy positions this novel combination therapy as an attractive strategy ready for clinical testing against a range of histologies, including RCC and NSCLC. Clin Cancer Res; 22(23); 5839-50. ©2016 AACR.


Asunto(s)
Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/terapia , Indoles/farmacología , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/terapia , Virus Oncolíticos/inmunología , Pirroles/farmacología , Reoviridae/inmunología , Inmunidad Adaptativa/inmunología , Animales , Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/virología , Línea Celular Tumoral , Terapia Combinada/métodos , Humanos , Neoplasias Renales/inmunología , Neoplasias Renales/virología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Viroterapia Oncolítica/métodos , Sunitinib , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
4.
Blood ; 118(24): 6368-79, 2011 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-21917757

RESUMEN

Chromosomal instability is a defining feature of clonal myeloma plasma cells that results in the perpetual accumulation of genomic aberrations. In addition to its role in protein homeostasis, the ubiquitin-proteasome system is also involved in the regulation of DNA damage-repair proteins. In the present study, we show that proteasome inhibition induces a "BRCAness" state in myeloma cells (MM), with depletion of their nuclear pool of ubiquitin and abrogation of H2AX polyubiquitylation, an essential step for the recruitment of BRCA1 and RAD51 to the sites of DNA double-stranded breaks (DSBs) and the initiation of homologous recombination (HR)-mediated DNA repair. Inhibition of poly-ADP-ribose-polymerase 1 and 2 (PARP1/2) with ABT-888 induced transient DNA DSBs that were rapidly resolved and thus had no effect on viability of the MM cells. In contrast, cotreatment of MM cell lines and primary CD138(+) cells with bortezomib and ABT-888 resulted in the sustained accumulation of unrepaired DNA DSBs with persistence of unubiquitylated γH2AX foci, lack of recruitment of BRCA1 and RAD51, and ensuing MM-cell death. The heightened cytotoxicity of ABT-888 in combination with bortezomib compared with either drug alone was also confirmed in MM xenografts in SCID mice. Our studies indicate that bortezomib impairs HR in MM and results in a contextual synthetic lethality when combined with PARP inhibitors.


Asunto(s)
Antineoplásicos/farmacología , Ácidos Borónicos/farmacología , Inhibidores Enzimáticos/farmacología , Mieloma Múltiple/tratamiento farmacológico , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Inhibidores de Proteasoma , Pirazinas/farmacología , Adulto , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Ácidos Borónicos/administración & dosificación , Ácidos Borónicos/efectos adversos , Ácidos Borónicos/uso terapéutico , Bortezomib , Línea Celular Tumoral , Células Cultivadas , Reparación del ADN/efectos de los fármacos , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/efectos adversos , Inhibidores Enzimáticos/uso terapéutico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones SCID , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Complejo de la Endopetidasa Proteasomal , Pirazinas/administración & dosificación , Pirazinas/efectos adversos , Pirazinas/uso terapéutico , ARN Mensajero/metabolismo , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Análisis de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Blood ; 117(23): 6202-13, 2011 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-21474670

RESUMEN

Integrin-ß7 (ITGB7) mRNA is detected in multiple myeloma (MM) cells and its presence is correlated with MAF gene activation. Although the involvement of several integrin family members in MM-stoma cell interaction is well documented, the specific biologic functions regulated by integrin-ß7 in MM are largely unknown. Clinically, we have correlated integrin-ß7 expression in MM with poor survival outcomes post autologous stem cell transplantation and postsalvage therapy with bortezomib. Functionally, we have found that shRNA-mediated silencing of ITGB7 reduces MM-cell adhesion to extra-cellular matrix elements (fibronectin, E-cadherin) and reverses cell-adhesion-mediated drug resistance (CAM-DR) sensitizing them to bortezomib and melphalan. In addition, ITGB7 silencing abrogated MM-cell transwell migration in response to SDF1α gradients, reduced vessel density in xenografted tumors, and altered MM cells in vivo homing into the BM. Mechanistically, ITGB7 knockdown inhibited focal adhesion kinase (FAK) and Src phosphorylation, Rac1 activation, and SUMOylation, reduced VEGF production in MM-BM stem cell cocultures and attenuated p65-NF-κB activity. Our findings support a role for integrin-ß7 in MM-cell adhesion, migration, and BM homing, and pave the way for a novel therapeutic approach targeting this molecule.


Asunto(s)
Movimiento Celular , Cadenas beta de Integrinas/metabolismo , Mieloma Múltiple/metabolismo , Animales , Antineoplásicos Alquilantes/farmacología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Ácidos Borónicos/farmacología , Bortezomib , Cadherinas/genética , Cadherinas/metabolismo , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Línea Celular Tumoral , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Técnicas de Cocultivo , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Fibronectinas/genética , Fibronectinas/metabolismo , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Humanos , Cadenas beta de Integrinas/genética , Melfalán/farmacología , Ratones , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Mieloma Múltiple/terapia , Invasividad Neoplásica , Trasplante de Neoplasias , Fosforilación/efectos de los fármacos , Fosforilación/genética , Proteínas Proto-Oncogénicas c-maf/genética , Proteínas Proto-Oncogénicas c-maf/metabolismo , Pirazinas/farmacología , Trasplante de Células Madre , Sumoilación/efectos de los fármacos , Sumoilación/genética , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Trasplante Autólogo , Trasplante Heterólogo , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/genética , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
6.
J Immunol ; 183(3): 1705-14, 2009 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-19570832

RESUMEN

Intact tolerogenic mechanisms preclude effective immunity against tumors, as most tumor Ags differ little from normal host Ags. In contrast, when tolerance fails, the immune system becomes inappropriately activated against an autoantigen. We postulated that CD8(+) T cells activated during autoimmunity are capable of protecting against tumors that express the targeted autoantigen. To test this hypothesis, double-transgenic 8.3-NOD-RIPTAg mice were developed (where NOD is nonobese diabetic, RIP is rat insulin promoter, and TAg is large T Ag). In this model, individuals with the RIPTAg transgene develop insulinoma; those expressing a transgenic TCR (8.3-TCR) recognizing the islet-specific glucose 6 phosphatase catalytic subunit-related protein (IGRP) harbor a peripheral immune system dominated by diabetogenic CD8(+) T cells. Although tumor emergence was significantly slower in 8.3-NOD-RIPTAg mice compared with NOD-RIPTAg mice, all 8.3-NOD-RIPTAg mice eventually developed insulinoma. Tumor emergence was not secondary to clonal deletion or anergy. Ag loss and MHC down-regulation were not apparent. Endogenous 8.3-TCR CD8(+) T cells were recruited to the tumor site and proliferated upon arrival to the tumor, although they were notably absent from the central parts of more advanced tumors. These results demonstrate that a breakdown of tolerance capable of causing autoimmune disease is insufficient for effective tumor immunity. Alterations in the tumor microenvironment may inhibit efficient and comprehensive delivery of CD8(+) T cells to all regions of the tumor. These data suggest that any immunotherapeutic strategy for cancer must involve enhancement of a proinflammatory tumor microenvironment in addition to inhibition of tolerogenic mechanisms.


Asunto(s)
Autoantígenos/inmunología , Autoinmunidad , Linfocitos T CD8-positivos/inmunología , Insulinoma/inmunología , Animales , Antígenos de Neoplasias/inmunología , Diabetes Mellitus/inmunología , Tolerancia Inmunológica , Ratones , Ratones Transgénicos , Neoplasias/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA