Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Sci Transl Med ; 10(422)2018 01 03.
Article in English | MEDLINE | ID: mdl-29298869

ABSTRACT

Immune checkpoint inhibitors, including those targeting programmed cell death protein 1 (PD-1), are reshaping cancer therapeutic strategies. Evidence suggests, however, that tumor response and patient survival are determined by tumor programmed death ligand 1 (PD-L1) expression. We hypothesized that preconditioning of the tumor immune microenvironment using targeted, virus-mediated interferon (IFN) stimulation would up-regulate tumor PD-L1 protein expression and increase cytotoxic T cell infiltration, improving the efficacy of subsequent checkpoint blockade. Oncolytic viruses (OVs) represent a promising form of cancer immunotherapy. For brain tumors, almost all studies to date have used direct intralesional injection of OV, because of the largely untested belief that intravenous administration will not deliver virus to this site. We show, in a window-of-opportunity clinical study, that intravenous infusion of oncolytic human Orthoreovirus (referred to herein as reovirus) leads to infection of tumor cells subsequently resected as part of standard clinical care, both in high-grade glioma and in brain metastases, and increases cytotoxic T cell tumor infiltration relative to patients not treated with virus. We further show that reovirus up-regulates IFN-regulated gene expression, as well as the PD-1/PD-L1 axis in tumors, via an IFN-mediated mechanism. Finally, we show that addition of PD-1 blockade to reovirus enhances systemic therapy in a preclinical glioma model. These results support the development of combined systemic immunovirotherapy strategies for the treatment of both primary and secondary tumors in the brain.


Subject(s)
Brain Neoplasms/therapy , Oncolytic Viruses/pathogenicity , Animals , Glioma/therapy , Humans , Immunotherapy/methods , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/metabolism
2.
Oncotarget ; 5(9): 2807-19, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24798549

ABSTRACT

Reovirus is a double stranded RNA virus, with an intrinsic preference for replication in KRAS mutant cells. As 45% of human colorectal cancers (CRC) harbor KRAS mutations, we sought to investigate its efficacy in KRAS mutant CRC cells, and examine its impact in combination with the topoisimerase-1 inhibitor, irinotecan. Reovirus efficacy was examined in the KRAS mutant HCT116, and the isogenic KRAS WT Hke3 cell line, and in the non-malignant rat intestinal epithelial cell line. Apoptosis was determined by flow cytometry and TUNEL staining. Combination treatment with reovirus and irintoecan was investigated in 15 CRC cell lines, including the HCT116 p21 isogenic cell lines. Reovirus preferentially induced apoptosis in KRAS mutant HCT116 cells compared to its isogenic KRAS WT derivative, and in KRAS mutant IEC cells. Reovirus showed a greater degree of caspase 3 activation with PARP 1 cleavage, and preferential inhibition of p21 protein expression in KRAS mutant cells. Reovirus synergistically induced growth inhibition when combined with irinotecan. This synergy was lost upon p21 gene knock out. Reovirus preferentially induces apoptosis in KRAS mutant colon cancer cells. Reovirus and irinotecan combination therapy is synergistic, p21 mediated, and represents a novel potential treatment for patients with CRC.


Subject(s)
Antineoplastic Agents/therapeutic use , Camptothecin/analogs & derivatives , Colorectal Neoplasms/therapy , Mutation/genetics , Oncolytic Virotherapy , Proto-Oncogene Proteins/genetics , Reoviridae/physiology , ras Proteins/genetics , Animals , Apoptosis , Blotting, Western , Camptothecin/therapeutic use , Cell Cycle , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Combined Modality Therapy , Drug Synergism , Humans , Irinotecan , Microscopy, Electron, Scanning , Proto-Oncogene Proteins p21(ras) , Rats , Tumor Cells, Cultured
3.
Mod Pathol ; 25(10): 1333-44, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22699519

ABSTRACT

We analyzed the in situ molecular correlates of infection from cancer patients treated with reovirus. Melanoma, colorectal, and ovarian cancer samples from such patients showed variable infection of the cancer cells but not the intermingled benign cells. RT in situ PCR showed most cancer cells contained the viral genome with threefold less having productive viral infection as documented by either tubulin or reoviral protein co-expression. Productive infection in the cancer cells was strongly correlated with co-expression of p38 and caspase-3 as well as apoptosis-related death (P<0.001). The cancer cell apoptotic death was due to a marked viral-induced inhibition of microRNA-let-7d that, in turn, upregulated caspase-3 activity. In summary, reovirus shows a striking tropism to cancer cells in clinical samples. A rate-limiting factor of reovirus-induced cancer cell death is productive viral infection that operates via the marked reduction of microRNA-let-7d and concomitant elevated caspase-3 expression.


Subject(s)
Apoptosis , Colorectal Neoplasms/pathology , Melanoma/pathology , MicroRNAs/metabolism , Oncolytic Virotherapy/methods , Orthoreovirus, Mammalian/physiology , Ovarian Neoplasms/pathology , Skin Neoplasms/pathology , Caspase 3/metabolism , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/virology , Female , Humans , Melanoma/metabolism , Melanoma/virology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/virology , Reoviridae Infections/virology , Skin Neoplasms/metabolism , Skin Neoplasms/virology , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Mol Cancer ; 8: 47, 2009 Jul 14.
Article in English | MEDLINE | ID: mdl-19594950

ABSTRACT

BACKGROUND: Reovirus type 3 Dearing strain (ReoT3D) has an inherent propensity to preferentially infect and destroy cancer cells. The oncolytic activity of ReoT3D as a single agent has been demonstrated in vitro and in vivo against various cancers, including colon, pancreatic, ovarian and breast cancers. Its human safety and potential efficacy are currently being investigated in early clinical trials. In this study, we investigated the in vitro combination effects of ReoT3D and chemotherapeutic agents against human non-small cell lung cancer (NSCLC). RESULTS: ReoT3D alone exerted significant cytolytic activity in 7 of 9 NSCLC cell lines examined, with the 50% effective dose, defined as the initial virus dose to achieve 50% cell killing after 48 hours of infection, ranging from 1.46 +/- 0.12 approximately 2.68 +/- 0.25 (mean +/- SD) log10 pfu/cell. Chou-Talalay analysis of the combination of ReoT3D with cisplatin, gemcitabine, or vinblastine demonstrated strong synergistic effects on cell killing, but only in cell lines that were sensitive to these compounds. In contrast, the combination of ReoT3D and paclitaxel was invariably synergistic in all cell lines tested, regardless of their levels of sensitivity to either agent. Treatment of NSCLC cell lines with the ReoT3D-paclitaxel combination resulted in increased poly (ADP-ribose) polymerase cleavage and caspase activity compared to single therapy, indicating enhanced apoptosis induction in dually treated NSCLC cells. NSCLC cells treated with the ReoT3D-paclitaxel combination showed increased proportions of mitotic and apoptotic cells, and a more pronounced level of caspase-3 activation was demonstrated in mitotically arrested cells. CONCLUSION: These data suggest that the oncolytic activity of ReoT3D can be potentiated by taxanes and other chemotherapeutic agents, and that the ReoT3D-taxane combination most effectively achieves synergy through accelerated apoptosis triggered by prolonged mitotic arrest.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/therapy , Mammalian orthoreovirus 3/physiology , Oncolytic Virotherapy/methods , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/virology , Caspases/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Combined Modality Therapy , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Drug Synergism , Enzyme Activation/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/virology , Paclitaxel/administration & dosage , Poly(ADP-ribose) Polymerases/metabolism , Vinblastine/administration & dosage , Virion/physiology , ras Proteins/metabolism , Gemcitabine
5.
Cancer Res ; 63(2): 348-53, 2003 Jan 15.
Article in English | MEDLINE | ID: mdl-12543787

ABSTRACT

The human reovirus is an oncolytic virus that specifically targets cancer cells with an activated Ras pathway. Because it is replication competent and highly specific for cancer cells, this virus has the potential to be an effective antimetastatic cancer agent through remote site delivery. In this study, we exploited the ability of reovirus to replicate in murine cells to test the efficacy of this virus in eliminating distal and/or metastatic tumors in immune-competent mice. We found that i.v. therapy with reovirus not only inhibited metastatic tumor growth but also led to a significant improvement in animal survival. Combining i.v. reovirus treatment with immune suppression (cyclosporine A or anti-CD4/anti-CD8 antibodies) resulted in further reduction in tumor size and a considerable prolongation in survival, compared with viral therapy alone. Combined therapy was also effective in overcoming a preexisting immunity to reovirus (a common occurrence in humans and thus a potential impediment to oncolytic effectiveness) to induce metastatic tumor regression. This is the first study to use systemic delivery of an oncolytic agent in conjunction with immune-suppressive drugs to effectively prolong animal survival. Altogether, our results suggest that i.v. reovirus therapy may present a feasible, novel alternative in the treatment of metastatic cancer in humans.


Subject(s)
Neoplasms, Experimental/therapy , Neoplasms, Experimental/virology , Retroviridae/physiology , Animals , Carcinoma, Lewis Lung/therapy , Carcinoma, Lewis Lung/virology , Combined Modality Therapy , Cyclosporine/pharmacology , Cytopathogenic Effect, Viral , Disease Models, Animal , Female , Immunosuppressive Agents/pharmacology , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Lung Neoplasms/virology , Mammary Neoplasms, Experimental/therapy , Mammary Neoplasms, Experimental/virology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Retroviridae/immunology
6.
Hum Gene Ther ; 13(5): 641-52, 2002 Mar 20.
Article in English | MEDLINE | ID: mdl-11916487

ABSTRACT

We have previously shown that human reovirus replication is restricted to cells with an activated Ras pathway, and that reovirus could be used as an effective oncolytic agent against human glioblastoma xenografts. This study examines in more detail the feasibility of reovirus as a therapeutic for breast cancer, a subset of cancer in which direct activating mutations in the ras proto-oncogene are rare, and yet where unregulated stimulation of Ras signaling pathways is important in the pathogenesis of the disease. We demonstrate herein the efficient lysis of breast tumor-derived cell lines by the virus, whereas normal breast cells resist infection in vitro. In vivo studies of reovirus breast cancer therapy reveal that viral administration could cause tumor regression in an MDA-MB-435S mammary fat pad model in severe combined immunodeficient mice. Reovirus could also effect regression of tumors remote from the injection site in an MDA-MB-468 bilateral tumor model, raising the possibility of systemic therapy of breast cancer by the oncolytic agent. Finally, the ability of reovirus to act against primary breast tumor samples not propagated as cell lines was evaluated; we found that reovirus could indeed replicate in ex vivo surgical specimens. Overall, reovirus shows promise as a potential breast cancer therapeutic.


Subject(s)
Biological Therapy , Breast Neoplasms/therapy , Reoviridae/physiology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Death/genetics , Female , Genes, ras , Genetic Therapy , Humans , Mice , Mice, SCID , Neoplasm Transplantation , Proto-Oncogene Mas , Transplantation, Heterologous , Tumor Cells, Cultured , Virus Replication/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...