Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
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.
PLoS One ; 12(1): e0168233, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28099441

RESUMEN

BACKGROUND AND PURPOSE: Reovirus is a ubiquitous RNA virus that exploits aberrant signaling pathways for its replication. The oncolytic potential of reovirus against numerous cancers under pre-clinical/clinical conditions has been documented by us and others. Despite its proven clinical activity, the underlying mechanisms of reovirus oncolysis is still not well elucidated. If reovirus therapy is to be optimized for cancer, including breast cancer patients, it is imperative to understand the mechanisms of reovirus oncolysis, especially in treatment of resistant tumour. EXPERIMENTAL APPROACH AND RESULTS: In the present study global gene expression profiling was utilized as a preliminary roadmap to tease-out pivotal molecules involved in reovirus induced apoptosis in breast cancer. Reovirus treated HTB133 and MCF7 breast cancer cells revealed transcriptional alteration of a defined subset of apoptotic genes and members of the nuclear factor-kappa B (NF-kB) family and p53 upregulated modulator of apoptosis (PUMA) were prominent. Since NF-kB can paradoxically suppress or promote apoptosis in cancer, the significance of NF-kB in reovirus oncolysis of breast cancer was investigated. Real time PCR analysis indicated a 2.9-4.3 fold increase in NF-kB p65 message levels following reovirus infection of MCF7 and HTB133, respectively. Nuclear translocation of NF-kB p65 protein was also dramatically augmented post reovirus treatment and correlated with enhanced DNA binding. Pharmacologic inhibition of NF-kB lead to oncolytic protection and significant down regulation of PUMA message levels. PUMA down regulation using siRNA suppressed reovirus oncolysis via significantly repressed apoptosis in p53 mutant HTB133 cells. CONCLUSIONS: This study demonstrates for the first time that a prominent pathway of reovirus oncolysis of breast cancer is mediated through NF-kB and that PUMA upregulation is dependent on NF-kB activation. These findings represent potential therapeutic indicators of reovirus treatment in future clinical trials.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Neoplasias de la Mama/terapia , Viroterapia Oncolítica/métodos , Proteínas Proto-Oncogénicas/metabolismo , Reoviridae/fisiología , Factor de Transcripción ReIA/metabolismo , Apoptosis/fisiología , Proteínas Reguladoras de la Apoptosis/genética , Línea Celular Tumoral , Ensayo de Cambio de Movilidad Electroforética , Humanos , Células MCF-7 , Proteínas Proto-Oncogénicas/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Transducción de Señal/fisiología
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.
Autophagy ; 9(3): 413-4, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23322106

RESUMEN

Multiple myeloma (MM) is a clonal plasma cell malignancy that accounts for 10-15% of newly diagnosed hematological cancers. Although significant advances have been made in the treatment of MM the disease still remains incurable. The oncolytic potential of reovirus has previously been demonstrated by others and us and is currently in phase III clinical trials for solid tumors. In addition a phase I clinical trial has recently been initiated for MM. Despite the clinical activity, the mechanism(s) of cell death caused by reovirus in MM is yet not yet well elucidated. A comprehensive understanding of reovirus-mediated histology-specific cell death mechanisms is imperative if this therapeutic is to become a standard of care for patients. Previously we have shown that reovirus-mediated cell death of breast and prostate cancer is orchestrated via apoptosis. The present study demonstrates for the first time that in addition to inducing apoptosis reovirus also upregulates autophagy during oncolysis of MM.


Asunto(s)
Apoptosis , Autofagia , Regulación Neoplásica de la Expresión Génica , Mieloma Múltiple/terapia , Viroterapia Oncolítica/métodos , Reoviridae/fisiología , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Ensayos Clínicos como Asunto , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mieloma Múltiple/metabolismo
5.
Clin Cancer Res ; 18(18): 4962-72, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22761466

RESUMEN

PURPOSE: Despite the recent advances made in the treatment of multiple myeloma, the disease still remains incurable. The oncolytic potential of reovirus has previously been shown and is currently in phase III clinical trials for solid tumors. We tested the hypothesis that reovirus can successfully target human multiple myeloma in vitro, ex vivo, and in vivo without affecting human hematopoietic stem cell (HHSC) re-population/differentiation in a murine model that partially recapitulates human multiple myeloma. EXPERIMENTAL DESIGN: Human myeloma cell lines and ex vivo tumor specimens were exposed to reovirus and oncolysis and mechanisms of cell death were assessed. RPMI 8226(GFP+) cells were injected intravenously to non-obese diabetic/severe combined immune deficient (NOD/SCID) mice and treated with live reovirus (LV) or dead virus (DV). Multiple myeloma disease progression was evaluated via whole-body fluorescence and bone marrow infiltration. HHSCs exposed to LV/DV were injected to NOD/SCID mice and re-population/differentiation was monitored. RESULTS: A total of six of seven myeloma cell lines and five of seven patient tumor specimens exposed to reovirus showed significant in vitro sensitivity. Tumor response of multiple myeloma by LV, but not DV, was confirmed by comparison of total tumor weights (P = 0.05), and bone marrow infiltration (1/6, LV; 5/6, DV). Mice injected with LV- or DV-exposed HHSCs maintained in vivo re-population/lineage differentiation showing a lack of viral effect on the stem cell compartment. Reovirus oncolysis was mediated primarily by activation of the apoptotic pathways. CONCLUSIONS: The unique ability of reovirus to selectively kill multiple myeloma while sparing HHSCs places it as a promising systemic multiple myeloma therapeutic for clinical testing.


Asunto(s)
Mieloma Múltiple/terapia , Viroterapia Oncolítica , Virus Oncolíticos , Reoviridae , Animales , Apoptosis , Autofagia , Médula Ósea/patología , Línea Celular Tumoral , Efecto Citopatogénico Viral , Femenino , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/virología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Cancer Res ; 70(6): 2435-44, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20215509

RESUMEN

Reovirus is a nonattenuated double-stranded RNA virus that exploits aberrant signaling pathways allowing selective cytotoxicity against multiple cancer histologies. The use of reovirus as a potential treatment modality for prostate cancer has not previously been described, and in this study evidence of in vitro and in vivo activity against prostate cancer was seen both in preclinical models and in six patients. The human prostate carcinoma cell lines PC-3, LN-CaP, and DU-145 exposed to replication-competent reovirus showed evidence of infection as illustrated by viral protein synthesis, cytopathic effect, and release of viral progeny. This oncolytic effect was found to be manifested through apoptosis, as DNA fragmentation, Apo 2.7 expression, Annexin V binding, and poly(ADP-ribose) polymerase cleavage were observed in live reovirus-infected cells, but not in uninfected or dead virus-treated cells. In vivo, hind flank severe combined immunodeficient/nonobese diabetic murine xenograft showed reduction in tumor size when treated with even a single intratumoral injection of reovirus. Finally, intralesional reovirus injections into a cohort of six patients with clinically organ-confined prostate cancer resulted in minimal side effects and evidence of antitumor activity. Histologic analysis after prostatectomy found a significant CD8 T-cell infiltration within the reovirus-injected areas as well as evidence of increased caspase-3 activity. These findings suggest that reovirus therapy may provide a promising novel treatment for prostate cancer and also imply a possible role for viral immune targeting of tumor.


Asunto(s)
Orthoreovirus Mamífero 3/fisiología , Viroterapia Oncolítica/métodos , Neoplasias de la Próstata/terapia , Animales , Apoptosis/fisiología , Línea Celular Tumoral , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/virología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Cancer Res ; 65(21): 9982-9990, 2005 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-16267023

RESUMEN

Myxoma virus, a poxvirus previously considered rabbit specific, can replicate productively in a variety of human tumor cells in culture. The purpose of this study was to determine if there was efficacy or toxicities of this oncolytic virus against experimental models of human malignant gliomas in vitro, in vivo, and ex vivo in malignant glioma specimens. In vitro, the majority of glioma cell lines tested (7 of 8, 87.5%) were fully permissive for myxoma virus replication and killed by infection. In vivo, intracerebral (i.c.) myxoma virus inoculation was well tolerated and produced only minimal focal inflammatory changes at the site of viral inoculation. U87 and U251 orthotopic xenograft models were used to assess myxoma virus efficacy in vivo. A single intratumoral injection of myxoma virus dramatically prolonged median survival compared with treatment with UV-inactivated myxoma virus. Median survival was not reached in myxoma virus-treated groups versus 47.3 days (U87; P = 0.0002) and 50.7 days (U251; P = 0.0027) in UV-inactivated myxoma virus-treated groups. Most myxoma virus-treated animals (12 of 13, 92%) were alive and apparently "cured" when the experiment was finished (>130 days). Interestingly, we found a selective and long-lived myxoma virus infection in gliomas in vivo. This is the first demonstration of the oncolytic activity of myxoma virus in vivo. The nonpathogenic nature of myxoma virus outside of the rabbit host, its capacity to be genetically modified, its ability to produce a long-lived infection in human tumor cells, and the lack of preexisting antibodies in the human population suggest that myxoma virus may be an attractive oncolytic agent against human malignant glioma.


Asunto(s)
Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/virología , Glioma/terapia , Glioma/virología , Myxoma virus/fisiología , Viroterapia Oncolítica/métodos , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Desnudos , Células 3T3 NIH , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Clin Cancer Res ; 10(24): 8561-76, 2004 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-15623640

RESUMEN

PURPOSE: Human reovirus type 3 has been proposed to kill cancer cells with an activated Ras signaling pathway. The purpose of this study was to investigate the efficacy of reovirus in immunocompetent glioma animal models and safety/toxicity in immunocompetent animals, including nonhuman primates. EXPERIMENTAL DESIGN: Racine glioma cells 9L and RG2 were implanted s.c. or intracranially in Fisher 344 rats with or without reovirus antibodies, followed by treatment of reovirus. To study whether reovirus kills contralateral tumors in the brain and to determine viral distribution, we established an in situ dual tumor model followed by reovirus intratumoral inoculation only into the ipsilateral tumor. To evaluate neurotoxicity/safety of reovirus, Cynomolgus monkeys and immunocompetent rats were given intracranially with reovirus, and pathological examination and/or behavioral studies were done. Viral shedding and clinical biochemistry were systematically studied in monkeys. RESULTS: Intratumorally given reovirus significantly suppressed the growth of both s.c. and intracranially tumors and significantly prolonged survival. The presence of reovirus-neutralizing antibodies did not abort the reovirus' antitumor effect. Reovirus inhibited glioma growth intracranially in the ipsilateral but not the contralateral tumors; viral load in ipsilateral tumors was 15 to 330-fold higher than the contralateral tumors. No encephalitis or behavioral abnormalities were found in monkeys and rats given reovirus intracranially. No treatment-related clinical biochemistry changes or diffuse histopathological abnormality were found in monkeys inoculated intracranially with Good Manufacturing Practice prepared reovirus. Microscopic changes were confined to the region of viral inoculation and were dose related, suggesting reovirus intracranially was well tolerated in nonhuman primates. CONCLUSIONS: These data show the efficacy and safety of reovirus when it is used in the treatment of gliomas in immunocompetent hosts. Inoculation of reovirus into the brain of nonhuman primates did not produce significant toxicities.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Orthoreovirus Mamífero 3/fisiología , Animales , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/virología , Encefalitis/etiología , Encefalitis/patología , Femenino , Glioblastoma/patología , Glioblastoma/virología , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunoglobulina G , Hibridación in Situ , Macaca fascicularis , Masculino , Orthoreovirus Mamífero 3/aislamiento & purificación , Aprendizaje por Laberinto , Modelos Animales , Pruebas de Neutralización , Ratas , Ratas Endogámicas F344 , Ratas Desnudas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tasa de Supervivencia , Células Tumorales Cultivadas
9.
Cancer Res ; 63(12): 3162-72, 2003 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12810644

RESUMEN

Medulloblastoma (MB), the most common pediatric brain tumor, is a highly malignant disease with a 5-year survival rate of only 60%. Tumor cells invade surrounding tissue and disseminate through cerebral spinal fluid, making treatment difficult. Human reovirus type 3 exploits an activated Ras pathway in tumor cells to support productive infection as an oncolytic virus. Here, we examined the ability of human reovirus to kill MB cells lines and surgical specimens in vitro and inhibit tumor growth/metastases in vivo. Most human MB cell lines tested (five of seven = 71.4%), two MB cell lines derived from spontaneously arising tumors in Patched-1(+/-) mice (two of two = 100%) and three MB primary cultures derived from surgical specimens, were susceptible to reovirus infection. Reovirus was internalized and transcribed in both susceptible and resistant cell lines. However, viral protein synthesis was restricted to cell lines with higher levels of activated Ras, suggesting that Ras plays a critical role in reovirus oncolysis in MB. Using an in vivo Daoy orthotopic animal model, we found that a single i.t. injection of reovirus dramatically prolonged survival compared with controls (160 versus 70 days, respectively; P = 0.0003). Repeating this experiment with GFP-labeled Daoy cells and multiple i.t. administrations of reovirus, we again found prolonged survival and a dramatic reduction in spinal and leptomeningeal metastases (66.7% in control injections versus 0.0% in the live virus group). These data suggest that this oncolytic virus may be a potentially effective novel therapy against human MB. Its ability to reduce metastases to the spinal cord could allow a reduction in the dose/field of total neuroaxis cerebral-spinal radiotherapy currently used to treat/prevent cerebral spinal fluid dissemination.


Asunto(s)
Terapia Biológica , Neoplasias Cerebelosas/terapia , Orthoreovirus Mamífero 3/fisiología , Meduloblastoma/secundario , Neoplasias Meníngeas/secundario , Neoplasias de la Médula Espinal/secundario , Animales , Esquema de Medicación , Activación Enzimática , Factor 2 Eucariótico de Iniciación/antagonistas & inhibidores , Femenino , Genes Reporteros , Genes p53 , Proteínas Fluorescentes Verdes , Humanos , Inyecciones Espinales , Proteínas Luminiscentes/análisis , Proteínas Luminiscentes/genética , Meduloblastoma/prevención & control , Meduloblastoma/terapia , Neoplasias Meníngeas/prevención & control , Ratones , Ratones Desnudos , Proteínas de Neoplasias/fisiología , Proteínas Proto-Oncogénicas p21(ras)/fisiología , Transducción de Señal , Neoplasias de la Médula Espinal/prevención & control , Transcripción Genética , Células Tumorales Cultivadas , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto , eIF-2 Quinasa/antagonistas & inhibidores , eIF-2 Quinasa/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...