Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Future Virol ; 13(9): 671-680, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30416535

ABSTRACT

Oncolytic viruses (OVs) do more than simply infect and kill host cells. The accepted mechanism of action for OVs consists of a primary lytic phase and a subsequent antitumor and antiviral immune response. However, not all cells are subject to the direct effects of OV therapy, and it is becoming clear that OVs can also impact uninfected cells in the periphery. This review discusses the effects of OVs on uninfected neighboring cells, so-called bystander effects, and implications for OV therapies alone or in combination with other standard of care chemotherapy.

2.
Viruses ; 10(3)2018 03 15.
Article in English | MEDLINE | ID: mdl-29543735

ABSTRACT

High Mobility Group Box 1 (HMGB1) is a multifunctional protein that plays various roles in the processes of inflammation, cancer, and other diseases. Many reports document abundant HMGB1 release following infection with oncolytic viruses (OVs). Further, other groups including previous reports from our laboratory highlight the synergistic effects of OVs with chemotherapy drugs. Here, we show that virus-free supernatants have varying cytotoxic potential, and HMGB1 is actively secreted by two established fibroblast cell lines (NIH 3T3 and 3T6-Swiss albino) following HSV1716 infection in vitro. Further, pharmacologic inhibition or genetic knock-down of HMGB1 reveals a role for HMGB1 in viral restriction, the ability to modulate bystander cell proliferation, and drug sensitivity in 3T6 cells. These data further support the multifactorial role of HMGB1, and suggest it could be a target for modulating the efficacy of oncolytic virus therapies alone or in combination with other frontline cancer treatments.


Subject(s)
HMGB1 Protein/metabolism , Herpes Simplex/metabolism , Herpes Simplex/virology , Simplexvirus/physiology , Animals , Bystander Effect/drug effects , Cell Line , Cell Proliferation/drug effects , Culture Media, Conditioned/pharmacology , Gene Knockdown Techniques , HMGB1 Protein/genetics , Herpes Simplex/drug therapy , Humans , Mice , NIH 3T3 Cells , Oncolytic Viruses/physiology , Simplexvirus/drug effects , Virus Replication/drug effects
3.
Hum Gene Ther ; 25(12): 1062-84, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25274574

ABSTRACT

The 8th International Conference on Oncolytic Virus Therapeutics meeting was held from April 10-13, 2014, in Oxford, United Kingdom. It brought together experts in the field of oncolytics from Europe, Asia, Australasia, and the Americas and provided a unique opportunity to hear the latest research findings in oncolytic virotherapy. Presentations of recent work were delivered in an informal and intimate setting afforded by a small group of attendees and an exquisitely focused conference topic. Here we describe the oral presentations and enable the reader to share in the benefits of bringing together experts to share their findings.


Subject(s)
Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses , Europe , Humans , Neoplasms/genetics , Neoplasms/virology , Research
4.
J Hepatocell Carcinoma ; 1: 149-61, 2014.
Article in English | MEDLINE | ID: mdl-27508184

ABSTRACT

Oncolytic herpes simplex virus (HSV1716), lacking the neurovirulence factor ICP34.5, has highly selective replication competence for cancer cells and has been used in clinical studies of glioma, melanoma, head and neck squamous cell carcinoma, pediatric non-central nervous system solid tumors, and malignant pleural mesothelioma. To date, 88 patients have received HSV1716 and the virus is well tolerated, with selective replication in tumor cells and no spread to surrounding normal tissue. We assessed the potential value of HSV1716 in preclinical studies with two human hepatocellular carcinoma cell lines, HuH7 and HepG2-luc. HSV1716 displayed excellent replication kinetics in vitro in HepG2-luc cells, a cell line engineered to express luciferase, and virus-mediated cell killing correlated with loss of light emissions from the cells. In vivo, the HepG2-luc cells readily formed light-emitting xenografts that were easily visualized by an in vivo imaging system and efficiently eliminated by HSV1716 oncolysis after intratumoral injection. HSV1716 also demonstrated strong efficacy signals in subcutaneous HuH7 xenografts in nude mice after intravenous administration of virus. In the HuH7 model, the intravenously injected virus replicated prolifically immediately after efficient tumor localization, resulting in highly significant reductions in tumor growth and enhanced survival. Our preclinical results demonstrate excellent tumor uptake of HSV1716, with prolific replication and potent oncolysis. These observations warrant a clinical study of HSV1716 in hepatocellular carcinoma.

5.
Oncolytic Virother ; 2: 57-74, 2013.
Article in English | MEDLINE | ID: mdl-27512658

ABSTRACT

Oncolytic viruses are emerging as a potential new way of treating cancers. They are selectively replication-competent viruses that propagate only in actively dividing tumor cells but not in normal cells and, as a result, destroy the tumor cells by consequence of lytic infection. At least six different oncolytic herpes simplex viruses (oHSVs) have undergone clinical trials worldwide to date, and they have demonstrated an excellent safety profile and intimations of efficacy. The first pivotal Phase III trial with an oHSV, talimogene laherparepvec (T-Vec [OncoVex(GM-CSF)]), is almost complete, with extremely positive early results reported. Intuitively, therapeutically beneficial interactions between oHSV and chemotherapeutic and targeted therapeutic drugs would be limited as the virus requires actively dividing cells for maximum replication efficiency and most anticancer agents are cytotoxic or cytostatic. However, combinations of such agents display a range of responses, with antagonistic, additive, or, perhaps most surprisingly, synergistic enhancement of antitumor activity. When synergistic interactions in cancer cell killing are observed, chemotherapy dose reductions that achieve the same overall efficacy may be possible, resulting in a valuable reduction of adverse side effects. Therefore, the combination of an oHSV with "standard-of-care" drugs makes a logical and reasonable approach to improved therapy, and the addition of a targeted oncolytic therapy with "standard-of-care" drugs merits further investigation, both preclinically and in the clinic. Numerous publications report such studies of oncolytic HSV in combination with other drugs, and we review their findings here. Viral interactions with cellular hosts are complex and frequently involve intracellular signaling networks, thus creating diverse opportunities for synergistic or additive combinations with many anticancer drugs. We discuss potential mechanisms that may lead to synergistic interactions.

6.
J Nucl Med ; 53(4): 647-54, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22414636

ABSTRACT

UNLABELLED: Oncolytic herpes viruses show promise for cancer treatment. However, it is unlikely that they will fulfill their therapeutic potential when used as monotherapies. An alternative strategy is to use these viruses not only as oncolytic agents but also as a delivery mechanism of therapeutic transgenes to enhance tumor cell killing. The herpes simplex virus 1 deletion mutant HSV1716 is a conditionally replicating oncolytic virus that selectively replicates in and lyses dividing tumor cells. It has a proven safety profile in clinical trials and has demonstrated efficacy as a gene-delivery vehicle. To enhance its therapeutic potential, we have engineered HSV1716 to convey the noradrenaline transporter (NAT) gene (HSV1716/NAT), whose expression endows infected cells with the capacity to accumulate the noradrenaline analog metaiodobenzylguanidine (MIBG). Thus, the NAT gene-infected cells are susceptible to targeted radiotherapy using radiolabeled (131)I-MIBG, a strategy that has already shown promise for combined targeted radiotherapy-gene therapy in cancer cells after plasmid-mediated transfection. METHODS: We used HSV1716/NAT as a dual cell lysis-gene delivery vehicle for targeting the NAT transgene to human tumor xenografts in vivo. RESULTS: In tumor xenografts that did not express NAT, intratumoral or intravenous injection of HSV1716/NAT induced the capacity for active uptake of (131)I-MIBG. Administration of HSV1716/NAT and (131)I-MIBG resulted in decreased tumor growth and enhanced survival relative to injection of either agent alone. Efficacy was dependent on the scheduling of delivery of the 2 agents. CONCLUSION: These findings support a role for combination radiotherapy-gene therapy for cancer using HSV1716 expressing the NAT transgene and targeted radionuclide therapy.


Subject(s)
Gene Transfer Techniques , Glioma/genetics , Glioma/radiotherapy , Herpesvirus 1, Human/genetics , Molecular Targeted Therapy/methods , Oncolytic Viruses/genetics , 3-Iodobenzylguanidine/administration & dosage , 3-Iodobenzylguanidine/pharmacokinetics , 3-Iodobenzylguanidine/therapeutic use , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , Combined Modality Therapy , DNA, Recombinant/genetics , Glioma/metabolism , Glioma/pathology , Humans , Mice , Norepinephrine Plasma Membrane Transport Proteins/genetics , Sequence Deletion , Time Factors
7.
Anticancer Res ; 29(6): 2159-66, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19528476

ABSTRACT

BACKGROUND: HSV1790 is an oncolytic virus generated by inserting the enzyme nitroreductase (NTR) into the virus HSV1716. NTR converts the prodrug CB1954 into an active alkylating agent. MATERIALS AND METHODS: In vitro, 3T6 cells (non permissive to HSV) were used in order to distinguish between virus-induced cytopathic effect and cell death due to activated prodrug. In vivo, xenograft models were injected with HSV1790 (10(5)-10(9) PFU) with or without CB1954 (max 80mg/kg) and tumor volume recorded regularly. Biodistribution of HSV1790 was determined immunohistochemically and by PCR. RESULTS: HSV1790 + CB1954 in vitro was more effective at killing tumor cells than the virus or the prodrug alone. In vivo, the combination reduced tumor volume and increased survival compared to treatment with HSV1790 or CB1954 alone. Following systemic administration of HSV1790, viral replication was detected in tumors, but not organs. CONCLUSION: HSV1790 + prodrug enhances tumor cell killing in vitro and reduces tumor volume and increases survival in vivo.


Subject(s)
Antineoplastic Agents/therapeutic use , Aziridines/therapeutic use , Herpesvirus 1, Human/pathogenicity , Neoplasms, Experimental/therapy , Oncolytic Viruses/metabolism , Prodrugs/therapeutic use , Animals , Blotting, Western , Combined Modality Therapy , Female , Herpes Simplex/genetics , Herpes Simplex/pathology , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Humans , Immunoenzyme Techniques , Mice , Mice, Nude , Neoplasms, Experimental/genetics , Neoplasms, Experimental/virology , Oncolytic Viruses/genetics , Polymerase Chain Reaction , Prodrugs/pharmacokinetics , Tissue Distribution , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...