51
|
To Infection and Beyond: The Multi-Pronged Anti-Cancer Mechanisms of Oncolytic Viruses. Viruses 2016; 8:v8020043. [PMID: 26861381 PMCID: PMC4776198 DOI: 10.3390/v8020043] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 01/17/2016] [Accepted: 02/01/2016] [Indexed: 12/12/2022] Open
Abstract
Over the past 1–2 decades we have witnessed a resurgence of efforts to therapeutically exploit the attributes of lytic viruses to infect and kill tumor cells while sparing normal cells. We now appreciate that the utility of viruses for treating cancer extends far beyond lytic cell death. Viruses are also capable of eliciting humoral and cellular innate and adaptive immune responses that may be directed not only at virus-infected cells but also at uninfected cancer cells. Here we review our current understanding of this bystander effect, and divide the mechanisms into lytic, cytokine, innate cellular, and adaptive phases. Knowing the key pathways and molecular players during virus infection in the context of the cancer microenvironment will be critical to devise strategies to maximize the therapeutic effects of oncolytic viroimmunotherapy.
Collapse
|
52
|
Fu X, Rivera A, Tao L, Zhang X. An HSV-2 based oncolytic virus can function as an attractant to guide migration of adoptively transferred T cells to tumor sites. Oncotarget 2015; 6:902-14. [PMID: 25460506 PMCID: PMC4359264 DOI: 10.18632/oncotarget.2817] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 11/24/2014] [Indexed: 11/25/2022] Open
Abstract
Adoptive T-cell therapy has shown promises for cancer treatment. However, for treating solid tumors, there is a need for improving the ability of the adoptively transferred T cells to home to tumor sites. We explored the possibility of using an oncolytic virus derived from HSV-2, which can actively pull T effector cells to the site of infection, as a local attractant for migration of adoptively transferred T cells. Our data show that intratumoral administration of this virus can indeed attract active migration of the adoptively transferred T cells to the treated tumor. Moreover, once attracted to the tumor site by the virus, T cells persisted in there significantly longer than in mock-treated tumor. Chemokine profiling identified significant elevation of CXCL9 and CXCL10, as well as several other chemokines belonging to the inflammatory chemokine family in the virus-treated tumors. These chemokines initially guided the T-cell migration to and then maintained their persistence in the tumor site, leading to a significantly enhanced therapeutic effect. Our data suggests that this virotherapy may be combined with adoptive T-cell therapy to potentiate its therapeutic effect against solid tumors that are otherwise difficult to manage with the treatment alone.
Collapse
Affiliation(s)
- Xinping Fu
- Department of Biology and Biochemistry and Center for Nuclear Receptors and Cell Signaling, University of Houston, Texas, USA
| | - Armando Rivera
- Department of Biology and Biochemistry and Center for Nuclear Receptors and Cell Signaling, University of Houston, Texas, USA
| | - Lihua Tao
- Department of Biology and Biochemistry and Center for Nuclear Receptors and Cell Signaling, University of Houston, Texas, USA
| | - Xiaoliu Zhang
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| |
Collapse
|
53
|
Pediatric cancer gone viral. Part I: strategies for utilizing oncolytic herpes simplex virus-1 in children. MOLECULAR THERAPY-ONCOLYTICS 2015; 2:S2372-7705(16)30017-1. [PMID: 26436135 PMCID: PMC4589755 DOI: 10.1038/mto.2015.15] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Progress for improving outcomes in pediatric patients with solid tumors remains slow. In addition, currently available therapies are fraught with numerous side effects, often causing significant life-long morbidity for long-term survivors. The use of viruses to kill tumor cells based on their increased vulnerability to infection is gaining traction, with several viruses moving through early and advanced phase clinical testing. The prospect of increased efficacy and decreased toxicity with these agents is thus attractive for pediatric cancer. In part I of this two-part review, we focus on strategies for utilizing oncolytic engineered herpes simplex virus (HSV) to target pediatric malignancies. We discuss mechanisms of action, routes of delivery, and the role of preexisting immunity on antitumor efficacy. Challenges to maximizing oncolytic HSV in children are examined, and we highlight how these may be overcome through various arming strategies. We review the preclinical and clinical evidence demonstrating safety of a variety of oncolytic HSVs. In Part II, we focus on the antitumor efficacy of oncolytic HSV in pediatric tumor types, pediatric clinical advances made to date, and future prospects for utilizing HSV in pediatric patients with solid tumors.
Collapse
|
54
|
Appleton ES, Turnbull S, Ralph C, West E, Scott K, Harrington K, Pandha H, Melcher A. Talimogene laherparepvec in the treatment of melanoma. Expert Opin Biol Ther 2015; 15:1517-30. [DOI: 10.1517/14712598.2015.1084280] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
55
|
Peters C, Rabkin SD. Designing Herpes Viruses as Oncolytics. MOLECULAR THERAPY-ONCOLYTICS 2015; 2:S2372-7705(16)30012-2. [PMID: 26462293 PMCID: PMC4599707 DOI: 10.1038/mto.2015.10] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Oncolytic herpes simplex virus (oHSV) was one of the first genetically-engineered oncolytic viruses. Because herpes simplex virus (HSV) is a natural human pathogen that can cause serious disease, it is incumbent that it be genetically-engineered or significantly attenuated for safety. Here we present a detailed explanation of the functions of HSV-1 genes frequently mutated to endow oncolytic activity. These genes are non-essential for growth in tissue culture cells but are important for growth in post-mitotic cells, interfering with intrinsic antiviral and innate immune responses or causing pathology, functions dispensable for replication in cancer cells. Understanding the function of these genes leads to informed creation of new oHSVs with better therapeutic efficacy. Virus infection and replication can also be directed to cancer cells through tumor-selective receptor binding and transcriptional- or post-transcriptional miRNA-targeting, respectively. In addition to the direct effects of oHSV on infected cancer cells and tumors, oHSV can be 'armed' with transgenes that are: reporters, to track virus replication and spread; cytotoxic, to kill uninfected tumor cells; immune modulatory, to stimulate anti-tumor immunity; or tumor microenvironment altering, to enhance virus spread or to inhibit tumor growth. In addition to HSV-1, other alphaherpesviruses are also discussed for their oncolytic activity.
Collapse
Affiliation(s)
- Cole Peters
- Program in Virology, Harvard Medical School, Boston, MA, and Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston MA
| | - Samuel D Rabkin
- Program in Virology, Harvard Medical School, Boston, MA, and Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston MA
| |
Collapse
|
56
|
Rajasekaran N, Chester C, Yonezawa A, Zhao X, Kohrt HE. Enhancement of antibody-dependent cell mediated cytotoxicity: a new era in cancer treatment. Immunotargets Ther 2015; 4:91-100. [PMID: 27471715 PMCID: PMC4918249 DOI: 10.2147/itt.s61292] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The therapeutic efficacy of some anti-tumor monoclonal antibodies (mAbs) depends on the capacity of the mAb to recognize the tumor-associated antigen and induce cytotoxicity via a network of immune effector cells. This process of antibody-dependent cell-mediated cytotoxicity (ADCC) against tumor cells is triggered by the interaction of the fragment crystallizable (Fc) portion of the mAb with the Fc receptors on effector cells like natural killer cells, macrophages, γδ T cells, and dendritic cells. By augmenting ADCC, the antitumor activity of mAbs can be significantly increased. Currently, identifying and developing therapeutic agents that enhance ADCC is a growing area of research. Combining existing tumor-targeting mAbs and ADCC-promoting agents that stimulate effector cells will translate to greater clinical responses. In this review, we discuss strategies for enhancing ADCC and emphasize the potential of combination treatments that include US Food and Drug Administration-approved mAbs and immunostimulatory therapeutics.
Collapse
Affiliation(s)
- Narendiran Rajasekaran
- Division of Oncology, Stanford School of Medicine, Stanford University, Stanford, CA, USA
| | - Cariad Chester
- Division of Oncology, Stanford School of Medicine, Stanford University, Stanford, CA, USA
| | - Atsushi Yonezawa
- Division of Oncology, Stanford School of Medicine, Stanford University, Stanford, CA, USA
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
| | - Xing Zhao
- Division of Oncology, Stanford School of Medicine, Stanford University, Stanford, CA, USA
- Tissue Engineering and Stem Cells Research Center, Department of Immunology, Guiyang Medical University, Guiyang, Guizhou Province, People’s Republic of China
| | - Holbrook E Kohrt
- Division of Oncology, Stanford School of Medicine, Stanford University, Stanford, CA, USA
| |
Collapse
|
57
|
Abstract
INTRODUCTION The clinical outcomes of patients with pancreatic cancer are poor, and the limited success of classical chemotherapy underscores the need for new, targeted approaches for this disease. The delivery of genetic material to cells allows for a variety of therapeutic concepts. Engineered agents based on synthetic biology are under clinical investigation in various cancers, including pancreatic cancer. AREAS COVERED This review focuses on Phase I - III clinical trials of gene and cell therapy for pancreatic cancer and on future implications of recent translational research. Trials available in the US National Library of Medicine (www.clinicaltrials.gov) until February 2014 were reviewed and relevant published results of preclinical and clinical studies were retrieved from www.pubmed.gov . EXPERT OPINION In pancreatic cancer, gene and cell therapies are feasible and may have synergistic antitumor activity with standard treatment and/or immunotherapy. Challenges are related to application safety, manufacturing costs, and a new spectrum of adverse events. Further studies are needed to evaluate available agents in carefully designed protocols and combination regimens. Enabling personalized cancer therapy, insights from molecular diagnostic technologies will guide the development and selection of new gene-based drugs. The evolving preclinical and clinical data on gene-based therapies can lay the foundation for future avenues improving patient care in pancreatic cancer.
Collapse
Affiliation(s)
- Hans Martin Singh
- National Center for Tumor Diseases and German Cancer Research Center, Department of Translational Oncology , Heidelberg , Germany
| | | | | |
Collapse
|
58
|
Abstract
Oncolytic viruses (OV) selectively replicate and kill cancer cells and spread within the tumor, while not harming normal tissue. In addition to this direct oncolytic activity, OVs are also very effective at inducing immune responses to themselves and to the infected tumor cells. OVs encompass a broad diversity of DNA and RNA viruses that are naturally cancer selective or can be genetically engineered. OVs provide a diverse platform for immunotherapy; they act as in situ vaccines and can be armed with immunomodulatory transgenes or combined with other immunotherapies. However, the interactions of OVs with the immune system may affect therapeutic outcomes in opposing fashions: negatively by limiting virus replication and/or spread, or positively by inducing antitumor immune responses. Many aspects of the OV-tumor/host interaction are important in delineating the effectiveness of therapy: (i) innate immune responses and the degree of inflammation induced; (ii) types of virus-induced cell death; (iii) inherent tumor physiology, such as infiltrating and resident immune cells, vascularity/hypoxia, lymphatics, and stromal architecture; and (iv) tumor cell phenotype, including alterations in IFN signaling, oncogenic pathways, cell surface immune markers [MHC, costimulatory, and natural killer (NK) receptors], and the expression of immunosuppressive factors. Recent clinical trials with a variety of OVs, especially those expressing granulocyte macrophage colony-stimulating factor (GM-CSF), have demonstrated efficacy and induction of antitumor immune responses in the absence of significant toxicity. Manipulating the balance between antivirus and antitumor responses, often involving overlapping immune pathways, will be critical to the clinical success of OVs.
Collapse
Affiliation(s)
- E Antonio Chiocca
- Authors' Affiliations: Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston Massachusetts
| | | |
Collapse
|
59
|
Wang J, Xu L, Zeng W, Hu P, Zeng M, Rabkin SD, Liu R. Treatment of human hepatocellular carcinoma by the oncolytic herpes simplex virus G47delta. Cancer Cell Int 2014; 14:83. [PMID: 25360068 PMCID: PMC4213511 DOI: 10.1186/s12935-014-0083-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 08/05/2014] [Indexed: 12/31/2022] Open
Abstract
Background Oncolytic herpes simplex virus (HSV) can replicate in and kill cancer cells while sparing the adjacent normal tissue. Hepatocellular carcinoma (HCC) is amongst the most common and lethal cancers, especially in Third World countries. In this study, the cytotoxicity of a third-generation oncolytic HSV, G47Δ, was investigated in different human HCC cell lines and in an immortalized human hepatic cell line. Additionally, subcutaneous models of HCC were established to evaluate the in vivo anti-tumor efficacy of G47Δ. Methods The HepG2, HepB, SMMC-7721, BEL-7404, and BEL-7405 human HCC cell lines and the HL-7702 human hepatic immortalized cell lines were infected with G47Δ at different multiplicities of infection (MOIs). The viability of infected cells was determined, and the G47Δ replication was identified by X-gal staining for LacZ expression. Two subcutaneous (s.c.) HCC tumor models of HCC were also established in Balb/c nude mice, which were intratumorally(i.t.) treated with either G47Δ or mock virus. Tumor volume and mouse survival times were documented. Results More than 95% of the HepG2, Hep3B,and SMMC-7721 HCC cells were killed on by day 5 after infection with a MOI’s of 0.01. For the HL-7702 human hepatic immortalized cells, 100% of the cells were killed on by day 5 after infection with a MOI’s of 0.01. The BEL-7404 HCC cell line was less susceptible with about 70% cells were killed by day 5 after infection with a MOI’s of 0.01. Whereas the BEL-7405 HCC cells were the least susceptible, with only 30% of the cells were killed. Both the SMMC-7721 and BEL-7404 cells form aggressive sc tumor models. G47Δ replicates in the tumors, such that most of the tumors regressed after the G47Δ-treatment, and treated tumor-bearing mice survived much longer than the control animals. Conclusions G47Δ effectively kills human HCC cells and an immortalized hepatic cell line at low MOI. Intra-tumor injection of G47Δ can induce a therapeutic effect and prolong the survival of treated mice bearing SMMC-7721 and BEL-7404 subcutaneously (s.c.) tumors. Thus, G47Δ may be useful as a novel therapeutic agent for HCC.
Collapse
Affiliation(s)
- Jiani Wang
- Breast Cancer Center, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, 510630 Guangzhou, China
| | - Lihua Xu
- Department of Oncology and Hematology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Weigen Zeng
- Breast Cancer Center, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, 510630 Guangzhou, China ; Department of Colorectal Surgery, Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, 17 Panjiayuan Nanli, Chaoyang District, 100021 Beijing, China
| | - Pan Hu
- Breast Cancer Center, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, 510630 Guangzhou, China
| | - Musheng Zeng
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Centre, Guangzhou, China
| | - Samuel D Rabkin
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, USA
| | - Renbin Liu
- Breast Cancer Center, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, 510630 Guangzhou, China
| |
Collapse
|
60
|
Antoszczyk S, Spyra M, Mautner VF, Kurtz A, Stemmer-Rachamimov AO, Martuza RL, Rabkin SD. Treatment of orthotopic malignant peripheral nerve sheath tumors with oncolytic herpes simplex virus. Neuro Oncol 2014; 16:1057-66. [PMID: 24470552 PMCID: PMC4096170 DOI: 10.1093/neuonc/not317] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUNDS Malignant peripheral nerve sheath tumors (MPNSTs) are an aggressive and often lethal sarcoma that frequently develops in patients with neurofibromatosis type 1 (NF1). We developed new preclinical MPNST models and tested the efficacy of oncolytic herpes simplex viruses (oHSVs), a promising cancer therapeutic that selectively replicates in and kills cancer cells. METHODS Mouse NF1(-) MPNST cell lines and human NF1(-) MPNST stemlike cells (MSLCs) were implanted into the sciatic nerves of immunocompetent and athymic mice, respectively. Tumor growth was followed by external measurement and sciatic nerve deficit using a hind-limb scoring system. Oncolytic HSV G47Δ as well as "armed" G47Δ expressing platelet factor 4 (PF4) or interleukin (IL)-12 were injected intratumorally into established sciatic nerve tumors. RESULTS Mouse MPNST cell lines formed tumors with varying growth kinetics. A single intratumoral injection of G47Δ in sciatic nerve tumors derived from human S462 MSLCs in athymic mice or mouse M2 (37-3-18-4) cells in immunocompetent mice significantly inhibited tumor growth and prolonged survival. Local IL-12 expression significantly improved the efficacy of G47Δ in syngeneic mice, while PF4 expression prolonged survival. Injection of G47Δ directly into the sciatic nerve of athymic mice resulted in only mild symptoms that did not differ from phosphate buffered saline control. CONCLUSIONS Two new orthotopic MPNST models are described, including in syngeneic mice, expanding the options for preclinical testing. Oncolytic HSV G47Δ exhibited robust efficacy in both immunodeficient and immunocompetent MPNST models while maintaining safety. Interleukin-12 expression improved efficacy. These studies support the clinical translation of G47Δ for patients with MPNST.
Collapse
Affiliation(s)
- Slawomir Antoszczyk
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts (S.A., R.L.M., S.D.R.); Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts (A.O.S.R.); Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.S., V.F.M.); Berlin-Brandenburg Center for Regenerative Therapies, Charité Medical University, Berlin, Germany (A.K.); College of Veterinary Medicine, Seoul National University, Seoul, Korea (A.K.)
| | - Melanie Spyra
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts (S.A., R.L.M., S.D.R.); Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts (A.O.S.R.); Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.S., V.F.M.); Berlin-Brandenburg Center for Regenerative Therapies, Charité Medical University, Berlin, Germany (A.K.); College of Veterinary Medicine, Seoul National University, Seoul, Korea (A.K.)
| | - Victor Felix Mautner
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts (S.A., R.L.M., S.D.R.); Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts (A.O.S.R.); Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.S., V.F.M.); Berlin-Brandenburg Center for Regenerative Therapies, Charité Medical University, Berlin, Germany (A.K.); College of Veterinary Medicine, Seoul National University, Seoul, Korea (A.K.)
| | - Andreas Kurtz
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts (S.A., R.L.M., S.D.R.); Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts (A.O.S.R.); Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.S., V.F.M.); Berlin-Brandenburg Center for Regenerative Therapies, Charité Medical University, Berlin, Germany (A.K.); College of Veterinary Medicine, Seoul National University, Seoul, Korea (A.K.)
| | - Anat O Stemmer-Rachamimov
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts (S.A., R.L.M., S.D.R.); Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts (A.O.S.R.); Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.S., V.F.M.); Berlin-Brandenburg Center for Regenerative Therapies, Charité Medical University, Berlin, Germany (A.K.); College of Veterinary Medicine, Seoul National University, Seoul, Korea (A.K.)
| | - Robert L Martuza
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts (S.A., R.L.M., S.D.R.); Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts (A.O.S.R.); Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.S., V.F.M.); Berlin-Brandenburg Center for Regenerative Therapies, Charité Medical University, Berlin, Germany (A.K.); College of Veterinary Medicine, Seoul National University, Seoul, Korea (A.K.)
| | - Samuel D Rabkin
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts (S.A., R.L.M., S.D.R.); Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts (A.O.S.R.); Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.S., V.F.M.); Berlin-Brandenburg Center for Regenerative Therapies, Charité Medical University, Berlin, Germany (A.K.); College of Veterinary Medicine, Seoul National University, Seoul, Korea (A.K.)
| |
Collapse
|
61
|
Cuddington BP, Mossman KL. Identifying roadblocks to successful oncolytic virotherapy: what are they and how do we approach them? Future Virol 2014. [DOI: 10.2217/fvl.14.48] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Breanne P Cuddington
- Department of Pathology & Molecular Medicine, McMaster Immunology Research Centre, Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Karen L Mossman
- Department of Pathology & Molecular Medicine, McMaster Immunology Research Centre, Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
62
|
Ning J, Wakimoto H. Oncolytic herpes simplex virus-based strategies: toward a breakthrough in glioblastoma therapy. Front Microbiol 2014; 5:303. [PMID: 24999342 PMCID: PMC4064532 DOI: 10.3389/fmicb.2014.00303] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/03/2014] [Indexed: 12/12/2022] Open
Abstract
Oncolytic viruses (OV) are a class of antitumor agents that selectively kill tumor cells while sparing normal cells. Oncolytic herpes simplex virus (oHSV) has been investigated in clinical trials for patients with the malignant brain tumor glioblastoma for more than a decade. These clinical studies have shown the safety of oHSV administration to the human brain, however, therapeutic efficacy of oHSV as a single treatment remains unsatisfactory. Factors that could hamper the anti-glioblastoma efficacy of oHSV include: attenuated potency of oHSV due to deletion or mutation of viral genes involved in virulence, restricting viral replication and spread within the tumor; suboptimal oHSV delivery associated with intratumoral injection; virus infection-induced inflammatory and cellular immune responses which could inhibit oHSV replication and promote its clearance; lack of effective incorporation of oHSV into standard-of-care, and poor knowledge about the ability of oHSV to target glioblastoma stem cells (GSCs). In an attempt to address these issues, recent research efforts have been directed at: (1) design of new engineered viruses to enhance potency, (2) better understanding of the role of the cellular immunity elicited by oHSV infection of tumors, (3) combinatorial strategies with different antitumor agents with a mechanistic rationale, (4) “armed” viruses expressing therapeutic transgenes, (5) use of GSC-derived models in oHSV evaluation, and (6) combinations of these. In this review, we will describe the current status of oHSV clinical trials for glioblastoma, and discuss recent research advances and future directions toward successful oHSV-based therapy of glioblastoma.
Collapse
Affiliation(s)
- Jianfang Ning
- Department of Neurosurgery, Brain Tumor Research Center, Massachusetts General Hospital, Harvard Medical School Boston, MA, USA
| | - Hiroaki Wakimoto
- Department of Neurosurgery, Brain Tumor Research Center, Massachusetts General Hospital, Harvard Medical School Boston, MA, USA
| |
Collapse
|
63
|
Gentschev I, Patil SS, Petrov I, Cappello J, Adelfinger M, Szalay AA. Oncolytic virotherapy of canine and feline cancer. Viruses 2014; 6:2122-37. [PMID: 24841386 PMCID: PMC4036544 DOI: 10.3390/v6052122] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 04/22/2014] [Accepted: 04/30/2014] [Indexed: 12/13/2022] Open
Abstract
Cancer is the leading cause of disease-related death in companion animals such as dogs and cats. Despite recent progress in the diagnosis and treatment of advanced canine and feline cancer, overall patient treatment outcome has not been substantially improved. Virotherapy using oncolytic viruses is one promising new strategy for cancer therapy. Oncolytic viruses (OVs) preferentially infect and lyse cancer cells, without causing excessive damage to surrounding healthy tissue, and initiate tumor-specific immunity. The current review describes the use of different oncolytic viruses for cancer therapy and their application to canine and feline cancer.
Collapse
Affiliation(s)
- Ivaylo Gentschev
- Department of Biochemistry, University of Wuerzburg, Wuerzburg D-97074, Germany.
| | - Sandeep S Patil
- Department of Biochemistry, University of Wuerzburg, Wuerzburg D-97074, Germany.
| | - Ivan Petrov
- Department of Biochemistry, University of Wuerzburg, Wuerzburg D-97074, Germany.
| | - Joseph Cappello
- Genelux Corporation, San Diego Science Center, San Diego, CA 92109, USA.
| | - Marion Adelfinger
- Department of Biochemistry, University of Wuerzburg, Wuerzburg D-97074, Germany.
| | - Aladar A Szalay
- Department of Biochemistry, University of Wuerzburg, Wuerzburg D-97074, Germany.
| |
Collapse
|
64
|
Bambury RM, Morris PG. The search for novel therapeutic strategies in the treatment of recurrent glioblastoma multiforme. Expert Rev Anticancer Ther 2014; 14:955-64. [PMID: 24814143 DOI: 10.1586/14737140.2014.916214] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive primary brain tumor with ≤10% patients surviving 5 years from the time of diagnosis. After tumor progression on frontline therapy with concomitant chemoradiotherapy followed by consolidation temozolomide there are few effective treatment options. Bevacizumab and nitrosureas are the most commonly used systemic options in this instance but no overall survival benefit has been demonstrated. In this review we outline the major avenues of research for treatment of recurrent GBM including anti-angiogenic, signaling pathway blockade and immunotherapy approaches. Results of recent trials as well as pertinent ongoing studies are discussed. Enrollment of patients to clinical trials as well as incorporation of correlative translational science studies to identify predictive biomarkers of treatment response will be key to improving outcomes in this devastating disease.
Collapse
Affiliation(s)
- Richard M Bambury
- Memorial Sloan-Kettering Cancer Center, 1275 York Avenue New York, NY 10065, USA
| | | |
Collapse
|
65
|
Zhao Q, Zhang W, Ning Z, Zhuang X, Lu H, Liang J, Li J, Zhang Y, Dong Y, Zhang Y, Zhang S, Liu S, Liu B. A novel oncolytic herpes simplex virus type 2 has potent anti-tumor activity. PLoS One 2014; 9:e93103. [PMID: 24671154 PMCID: PMC3966855 DOI: 10.1371/journal.pone.0093103] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 02/28/2014] [Indexed: 02/02/2023] Open
Abstract
Oncolytic viruses are promising treatments for many kinds of solid tumors. In this study, we constructed a novel oncolytic herpes simplex virus type 2: oHSV2. We investigated the cytopathic effects of oHSV2 in vitro and tested its antitumor efficacy in a 4T1 breast cancer model. We compared its effect on the cell cycle and its immunologic impact with the traditional chemotherapeutic agent doxorubicin. In vitro data showed that oHSV2 infected most of the human and murine tumor cell lines and was highly oncolytic. oHSV2 infected and killed 4T1 tumor cells independent of their cell cycle phase, whereas doxorubicin mainly blocked cells that were in S and G2/M phase. In vivo study showed that both oHSV2 and doxorubicin had an antitumor effect, though the former was less toxic. oHSV2 treatment alone not only slowed down the growth of tumors without causing weight loss but also induced an elevation of NK cells and mild decrease of Tregs in spleen. In addition, combination therapy of doxorubicin followed by oHSV2 increased survival with weight loss than oHSV2 alone. The data showed that the oncolytic activity of oHSV2 was similar to oHSV1 in cell lines examined and in vivo. Therefore, we concluded that our virus is a safe and effective therapeutic agent for 4T1 breast cancer and that the sequential use of doxorubicin followed by oHSV2 could improve antitumor activity without enhancing doxorubicin’s toxicity.
Collapse
Affiliation(s)
- Qian Zhao
- Department of Pathology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wen Zhang
- Department of Immunology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhifeng Ning
- School of Pharmacology, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Xiufen Zhuang
- Department of Immunology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Haizhen Lu
- Department of Pathology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jing Liang
- Department of Pathology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jie Li
- Department of Immunology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yu Zhang
- Department of Immunology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ying Dong
- Department of Immunology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Youhui Zhang
- Department of Immunology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shuren Zhang
- Department of Immunology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shangmei Liu
- Department of Pathology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Binlei Liu
- Department of Immunology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; School of Pharmacology, Hubei University of Science and Technology, Xianning, Hubei, China
| |
Collapse
|
66
|
Abstract
Cancer-permissive viruses or oncolytic viruses consist of either genetically engineered or naturally occurring strains that possess relatively selective replicative and/or infection abilities for cancer vs. normal cells (Chiocca, Nat Rev Cancer 2: 938-950, 2002). They can also be armed with additional anticancer cDNAs (e.g., cytokines, prodrug-activating, anti-angiogenesis genes, and others) to extend therapeutic effects (Kaur et al., Curr Gene Ther 9: 341-355, 2009). Herpes simplex virus type 1 (HSV-1) possesses several advantages as an oncolytic virus such as a rapid lytic cycle and a large capacity for insertion of heterologous DNA sequences (Wade-Martins et al., Nat Biotechnol, 19: 1067-1070, 2001). However, the technical nuances of genetic manipulation of the HSV-1 genome may still be relatively challenging. Here, we describe a system that has been durable and consistent in providing the ability to generate multiple recombinant HSV-1. The HsvQuik technology utilizes an HSV-1 genome cloned in a bacterial artificial chromosome to recombine heterologous cDNAs in a relatively rapid and reliable manner (Terada et al., Gene Ther 13: 705-714, 2006).
Collapse
Affiliation(s)
- Hiroshi Nakashima
- Harvey Cushing Neuro-oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital/Dana-Farber Cancer Institute and Harvard Medical School, PBB3, 75 Francis St., Boston, MA, 02115, USA
| | | |
Collapse
|
67
|
Gaston DC, Odom CI, Li L, Markert JM, Roth JC, Cassady KA, Whitley RJ, Parker JN. Production of bioactive soluble interleukin-15 in complex with interleukin-15 receptor alpha from a conditionally-replicating oncolytic HSV-1. PLoS One 2013; 8:e81768. [PMID: 24312353 PMCID: PMC3842420 DOI: 10.1371/journal.pone.0081768] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 10/16/2013] [Indexed: 12/27/2022] Open
Abstract
Oncolytic type-1 herpes simplex viruses (oHSVs) lacking the γ134.5 neurovirulence gene are being evaluated for treatment of a variety of malignancies. oHSVs replicate within and directly kill permissive cancer cells. To augment their anti-tumor activity, oHSVs have been engineered to express immunostimulatory molecules, including cytokines, to elicit tumor-specific immune responses. Interleukin-15 (IL-15) holds potential as an immunotherapeutic cytokine because it has been demonstrated to promote both natural killer (NK) cell-mediated and CD8(+) T cell-mediated cytotoxicity against cancer cells. The purpose of these studies was to engineer an oHSV producing bioactive IL-15. Two oHSVs were constructed encoding murine (m)IL-15 alone (J100) or with the mIL-15 receptor α (mIL-15Rα, J100D) to determine whether co-expression of these proteins is required for production of bioactive mIL-15 from oHSV. The following were demonstrated: i) both oHSVs retain replication competence and cytotoxicity in permissive tumor cell lines. ii) Enhanced production of mIL-15 was detected in cell lysates of neuro-2a cells following J100D infection as compared to J100 infection, suggesting that mIL-15Rα improved mIL-15 production. iii) Soluble mIL-15 in complex with mIL-15Rα was detected in supernates from J100D-infected, but not J100-infected, neuro-2a, GL261, and CT-2A cells. These cell lines vary in permissiveness to oHSV replication and cytotoxicity, demonstrating soluble mIL-15/IL-15Rα complex production from J100D was independent of direct oHSV effects. iv) The soluble mIL-15/IL-15Rα complex produced by J100D was bioactive, stimulating NK cells to proliferate and reduce the viability of syngeneic GL261 and CT-2A cells. v) J100 and J100D were aneurovirulent inasmuch as no neuropathologic effects were documented following direct inoculation into brains of CBA/J mice at up to 1x10(7) plaque forming units. The production of mIL-15/mIL-15Rα from multiple tumor lines, as well as the lack of neurovirulence, renders J100D suitable for investigating the combined effects of oHSV and mIL-15/IL-15Rα in various cancer models.
Collapse
Affiliation(s)
- David C Gaston
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America ; School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | | | | | | | | | | | | | | |
Collapse
|
68
|
Affiliation(s)
- Jianfang Ning
- Brain Tumor Research Center; Massachusetts General Hospital; Boston, MA USA
| | - Hiroaki Wakimoto
- Brain Tumor Research Center; Massachusetts General Hospital; Boston, MA USA
| | - Samuel D Rabkin
- Brain Tumor Research Center; Massachusetts General Hospital; Boston, MA USA
| |
Collapse
|
69
|
Immunogenic HSV-mediated oncolysis shapes the antitumor immune response and contributes to therapeutic efficacy. Mol Ther 2013; 22:123-31. [PMID: 24343053 DOI: 10.1038/mt.2013.238] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Accepted: 09/30/2013] [Indexed: 01/03/2023] Open
Abstract
Within the oncolytic virus field, the extent of virus replication that is essential for immune stimulation to control tumor growth remains unresolved. Using infected cell protein 0 (ICP0)-defective oncolytic Herpes simplex virus type 1 (HSV-1) and HSV-2 viruses (dICP0 and dNLS) that show differences in their in vitro replication and cytotoxicity, we investigated the inherent features of oncolytic HSV viruses that are required for potent antitumor activity. In vitro, the HSV-2 vectors showed rapid cytotoxicity despite lower viral burst sizes compared to HSV-1 vectors. In vivo, although both of the dICP0 vectors initially replicated to a similar level, HSV-1 dICP0 was rapidly cleared from the tumors. In spite of this rapid clearance, HSV-1 dICP0 treatment conferred significant survival benefit. HSV-1 dICP0-treated tumors showed significantly higher levels of danger-associated molecular patterns that correlated with higher numbers of antigen-presenting cells within the tumor and increased antigen-specific CD8+ T-cell levels in the peripheral blood. This study suggests that, at least in the context of oncolytic HSV, the initial stages of immunogenic virus replication leading to activation of antitumor immunity are more important than persistence of a replicating virus within the tumor. This knowledge provides important insight for the design of therapeutically successful oncolytic viruses.
Collapse
|
70
|
Bartlett DL, Liu Z, Sathaiah M, Ravindranathan R, Guo Z, He Y, Guo ZS. Oncolytic viruses as therapeutic cancer vaccines. Mol Cancer 2013; 12:103. [PMID: 24020520 PMCID: PMC3847443 DOI: 10.1186/1476-4598-12-103] [Citation(s) in RCA: 224] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 09/06/2013] [Indexed: 12/24/2022] Open
Abstract
Oncolytic viruses (OVs) are tumor-selective, multi-mechanistic antitumor agents. They kill infected cancer and associated endothelial cells via direct oncolysis, and uninfected cells via tumor vasculature targeting and bystander effect. Multimodal immunogenic cell death (ICD) together with autophagy often induced by OVs not only presents potent danger signals to dendritic cells but also efficiently cross-present tumor-associated antigens from cancer cells to dendritic cells to T cells to induce adaptive antitumor immunity. With this favorable immune backdrop, genetic engineering of OVs and rational combinations further potentiate OVs as cancer vaccines. OVs armed with GM-CSF (such as T-VEC and Pexa-Vec) or other immunostimulatory genes, induce potent anti-tumor immunity in both animal models and human patients. Combination with other immunotherapy regimens improve overall therapeutic efficacy. Coadministration with a HDAC inhibitor inhibits innate immunity transiently to promote infection and spread of OVs, and significantly enhances anti-tumor immunity and improves the therapeutic index. Local administration or OV mediated-expression of ligands for Toll-like receptors can rescue the function of tumor-infiltrating CD8+ T cells inhibited by the immunosuppressive tumor microenvironment and thus enhances the antitumor effect. Combination with cyclophosphamide further induces ICD, depletes Treg, and thus potentiates antitumor immunity. In summary, OVs properly armed or in rational combinations are potent therapeutic cancer vaccines.
Collapse
Affiliation(s)
- David L Bartlett
- University of Pittsburgh Cancer Institute and Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | | | | | | | | | | | | |
Collapse
|
71
|
Meshii N, Takahashi G, Okunaga S, Hamada M, Iwai S, Takasu A, Ogawa Y, Yura Y. Enhancement of systemic tumor immunity for squamous cell carcinoma cells by an oncolytic herpes simplex virus. Cancer Gene Ther 2013; 20:493-8. [PMID: 23887644 DOI: 10.1038/cgt.2013.45] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 06/07/2013] [Accepted: 06/15/2013] [Indexed: 01/13/2023]
Abstract
RH2 is a neurovirulent γ134.5 gene-deficient herpes simplex virus type 1 (HSV-1) with a lytic ability in human squamous cell carcinoma (SCC) cells; it is related to spontaneously occurring HSV-1 mutant HF10. The effect of RH2 on SCC was examined using a syngeneic C3H mouse model. After infection of mouse SCCVII cells with RH2, cell viability was decreased at first, but recovered by prolonged culture, indicating the limited replication of RH2. The antitumor ability of RH2 was examined using a bilateral SCCVII tumor model. The growth of the RH2-injected tumors was suppressed compared with that of phosphate-buffered saline-injected tumors. Moreover, the growth of contralateral tumor of RH2-treated mice was also suppressed significantly. The splenocytes of C3H mice treated with RH2 lysed more SCCVII cells than NFSaY83 cells and YAC-1 cells. The cytotoxicity of the splenocytes on SCCVII cells was significantly greater than that of splenocytes from tumor-bearing mice. Removal of CD8(+) T cells from splenocytes decreased their cell killing activity remarkably. The antitumor effect of RH2 on SCCVII xenografts in nude mice was not demonstrated. These results indicate that RH2 exhibited a suppressive effect on mouse SCC, even if the replication of RH2 was limited. This is ascribed to the ability of RH2 to enhance existing tumor-specific cytotoxic T lymphocyte activity.
Collapse
Affiliation(s)
- N Meshii
- Department of Oral and Maxillofacial Surgery, Osaka University Graduate School of Dentistry, Osaka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
72
|
Currier MA, Eshun FK, Sholl A, Chernoguz A, Crawford K, Divanovic S, Boon L, Goins WF, Frischer JS, Collins MH, Leddon JL, Baird WH, Haseley A, Streby KA, Wang PY, Hendrickson BW, Brekken RA, Kaur B, Hildeman D, Cripe TP. VEGF blockade enables oncolytic cancer virotherapy in part by modulating intratumoral myeloid cells. Mol Ther 2013; 21:1014-23. [PMID: 23481323 DOI: 10.1038/mt.2013.39] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Understanding the host response to oncolytic viruses is important to maximize their antitumor efficacy. Despite robust cytotoxicity and high virus production of an oncolytic herpes simplex virus (oHSV) in cultured human sarcoma cells, intratumoral (ITu) virus injection resulted in only mild antitumor effects in some xenograft models, prompting us to characterize the host inflammatory response. Virotherapy induced an acute neutrophilic infiltrate, a relative decrease of ITu macrophages, and a myeloid cell-dependent upregulation of host-derived vascular endothelial growth factor (VEGF). Anti-VEGF antibodies, bevacizumab and r84, the latter of which binds VEGF and selectively inhibits binding to VEGF receptor-2 (VEGFR2) but not VEGFR1, enhanced the antitumor effects of virotherapy, in part due to decreased angiogenesis but not increased virus production. Neither antibody affected neutrophilic infiltration but both partially mitigated virus-induced depletion of macrophages. Enhancement of virotherapy-mediated antitumor effects by anti-VEGF antibodies could largely be recapitulated by systemic depletion of CD11b(+) cells. These data suggest the combined effect of oHSV virotherapy and anti-VEGF antibodies is in part due to modulation of a host inflammatory reaction to virus. Our data provide strong preclinical support for combined oHSV and anti-VEGF antibody therapy and suggest that understanding and counteracting the innate host response may help enable the full antitumor potential of oncolytic virotherapy.
Collapse
Affiliation(s)
- Mark A Currier
- Divison of Oncology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Abstract
A common-cold virus uses an undercover strategy to avoid neutralizing antibodies in cancer patients and targets distant sites of metastatic cancer growth.
Collapse
Affiliation(s)
- John C Bell
- Center for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, and Department of Medicine, University of Ottawa, Ottawa, ON K1H 8L6, Canada.
| |
Collapse
|
74
|
Moralès O, Richard A, Martin N, Mrizak D, Sénéchal M, Miroux C, Pancré V, Rommelaere J, Caillet-Fauquet P, de Launoit Y, Delhem N. Activation of a helper and not regulatory human CD4+ T cell response by oncolytic H-1 parvovirus. PLoS One 2012; 7:e32197. [PMID: 22359669 PMCID: PMC3281136 DOI: 10.1371/journal.pone.0032197] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Accepted: 01/24/2012] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND H-1 parvovirus (H-1 PV), a rodent autonomous oncolytic parvovirus, has emerged as a novel class of promising anticancer agents, because of its ability to selectively find and destroy malignant cells. However, to probe H-1 PV multimodal antitumor potential one of the major prerequisites is to decipher H-1 PV direct interplay with human immune system, and so prevent any risk of impairment. METHODOLOGY/PRINCIPAL FINDINGS Non activated peripheral blood mononuclear cells (PBMCs) are not sensitive to H-1 PV cytotoxic effect. However, the virus impairs both activated PBMC proliferation ability and viability. This effect is related to H-1 PV infection as evidenced by Western blotting detection of H-1 PV main protein NS1. However, TCID50 experiments did not allow newly generated virions to be detected. Moreover, flow cytometry has shown that H-1 PV preferentially targets B lymphocytes. Despite seeming harmful at first sight, H-1 PV seems to affect very few NK cells and CD8+ T lymphocytes and, above all, clearly does not affect human neutrophils and one of the major CD4+ T lymphocyte subpopulation. Very interestingly, flow cytometry analysis and ELISA assays proved that it even activates human CD4+ T cells by increasing activation marker expression (CD69 and CD30) and both effective Th1 and Th2 cytokine secretion (IL-2, IFN-γ and IL-4). In addition, H-1 PV action does not come with any sign of immunosuppressive side effect. Finally, we have shown the efficiency of H-1 PV on xenotransplanted human nasopharyngeal carcinoma, in a SCID mouse model reconstituted with human PBMC. CONCLUSIONS/SIGNIFICANCE Our results show for the first time that a wild-type oncolytic virus impairs some immune cell subpopulations while directly activating a Helper CD4+ T cell response. Thus, our data open numerous gripping perspectives of investigation and strongly argue for the use of H-1 PV as an anticancer treatment.
Collapse
Affiliation(s)
- Olivier Moralès
- Institut de Biologie de Lille, UMR 8161, CNRS, Institut Pasteur de Lille, Université Lille-Nord de France, Lille, France
| | - Audrey Richard
- Institut de Biologie de Lille, UMR 8161, CNRS, Institut Pasteur de Lille, Université Lille-Nord de France, Lille, France
| | - Nathalie Martin
- Institut de Biologie de Lille, UMR 8161, CNRS, Institut Pasteur de Lille, Université Lille-Nord de France, Lille, France
| | - Dhafer Mrizak
- Institut de Biologie de Lille, UMR 8161, CNRS, Institut Pasteur de Lille, Université Lille-Nord de France, Lille, France
| | - Magalie Sénéchal
- Institut de Biologie de Lille, UMR 8161, CNRS, Institut Pasteur de Lille, Université Lille-Nord de France, Lille, France
| | - Céline Miroux
- Institut de Biologie de Lille, UMR 8161, CNRS, Institut Pasteur de Lille, Université Lille-Nord de France, Lille, France
| | - Véronique Pancré
- Institut de Biologie de Lille, UMR 8161, CNRS, Institut Pasteur de Lille, Université Lille-Nord de France, Lille, France
| | - Jean Rommelaere
- Tumor Virology, Research Program Infection and Cancer, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | - Perrine Caillet-Fauquet
- Institut de Biologie de Lille, UMR 8161, CNRS, Institut Pasteur de Lille, Université Lille-Nord de France, Lille, France
| | - Yvan de Launoit
- Institut de Biologie de Lille, UMR 8161, CNRS, Institut Pasteur de Lille, Université Lille-Nord de France, Lille, France
| | - Nadira Delhem
- Institut de Biologie de Lille, UMR 8161, CNRS, Institut Pasteur de Lille, Université Lille-Nord de France, Lille, France
- * E-mail:
| |
Collapse
|
75
|
Todo T. Active immunotherapy: oncolytic virus therapy using HSV-1. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 746:178-86. [PMID: 22639168 DOI: 10.1007/978-1-4614-3146-6_14] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Conditionally replicating herpes simplex viruses Type 1 (HSV-1) are promising therapeutic agents for glioma. They can replicate in situ, spread and exhibit oncolytic activity via a direct cytocidal effect. In addition, specific antitumor immunity is effectively induced in the course of oncolytic activities. G47Δ is a genetically engineered HSV-1 with triple mutations that realized augmented viral replication in tumor cells, strong induction of antitumor immunity and enhanced safety in normal tissues. A clinical trial of G47Δ in patients with recurrent glioblastoma has started in 2009. One of the advantages of HSV-1 is its capacity to incorporate large and/or multiple transgenes within the viral genome. In preclinical studies, "arming" of an oncolytic HSV-1 with transgenes encoding immunomodulatory molecules, such as interleukin 12, has been shown to greatly augment the efficacy of oncolytic HSV-1 therapy. Oncolytic virus therapy using HSV-1 may be a useful treatment for glioma that can also function as an efficient in situ tumor vaccination.
Collapse
Affiliation(s)
- Tomoki Todo
- Department of Neurosurgery, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
76
|
Deciphering the Multifaceted Relationship between Oncolytic Viruses and Natural Killer Cells. Adv Virol 2011; 2012:702839. [PMID: 22312364 PMCID: PMC3263705 DOI: 10.1155/2012/702839] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Accepted: 09/14/2011] [Indexed: 12/23/2022] Open
Abstract
Despite active research in virotherapy, this apparently safe modality has not achieved widespread success. The immune response to viral infection appears to be an essential factor that determines the efficacy of oncolytic viral therapy. The challenge is determining whether the viral-elicited immune response is a hindrance or a tool for viral treatment. NK cells are a key component of innate immunity that mediates antiviral immunity while also coordinating tumor clearance. Various reports have suggested that the NK response to oncolytic viral therapy is a critical factor in premature viral clearance while also mediating downstream antitumor immunity. As a result, particular attention should be given to the NK cell response to various oncolytic viral vectors and how their antiviral properties can be suppressed while maintaining tumor clearance. In this review we discuss the current literature on the NK response to oncolytic viral infection and how future studies clarify this intricate response.
Collapse
|
77
|
Takahashi S, Fusaki N, Ohta S, Iwahori Y, Iizuka Y, Inagawa K, Kawakami Y, Yoshida K, Toda M. Downregulation of KIF23 suppresses glioma proliferation. J Neurooncol 2011; 106:519-29. [PMID: 21904957 DOI: 10.1007/s11060-011-0706-2] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Accepted: 08/18/2011] [Indexed: 12/18/2022]
Abstract
To identify therapeutic molecular targets for glioma, we performed modified serological identification of antigens by recombinant complementary DNA (cDNA) expression cloning using sera from a mouse glioma model. Two clones, kinesin family member 23 (Kif23) and structural maintenance of chromosomes 4 (Smc4), were identified as antigens through immunological reaction with sera from mice harboring synergic GL261 mouse glioma and intratumoral inoculation with a mutant herpes simplex virus. The human Kif23 homolog KIF23 is a nuclear protein that localizes to the interzone of mitotic spindles, acting as a plus-end-directed motor enzyme that moves antiparallel microtubules in vitro. Expression analysis revealed a higher level of KIF23 expression in glioma tissues than in normal brain tissue. The introduction of small interfering RNA (siRNA) targeting KIF23 into two different glioma cell lines, U87MG and SF126, downregulated KIF23 expression, which significantly suppressed glioma cell proliferation in vitro. KIF23 siRNA-treated glioma cells exhibited larger cell bodies with two or more nuclei compared with control cells. In vivo analysis using mouse xenograft showed that KIF23 siRNA/DNA chimera-treated tumors were significantly smaller than tumors treated with control siRNA/DNA chimera. Taken together, our results indicate that downregulation of KIF23 decreases proliferation of glioma cells and that KIF23 may be a novel therapeutic target in malignant glioma.
Collapse
Affiliation(s)
- Satoshi Takahashi
- Department of Neurosurgery, Keio University, School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Schierer S, Hesse A, Knippertz I, Kaempgen E, Baur AS, Schuler G, Steinkasserer A, Nettelbeck DM. Human dendritic cells efficiently phagocytose adenoviral oncolysate but require additional stimulation to mature. Int J Cancer 2011; 130:1682-94. [PMID: 21557217 DOI: 10.1002/ijc.26176] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Accepted: 04/12/2011] [Indexed: 01/12/2023]
Abstract
Oncolytic adenoviruses are emerging agents for treatment of cancer by tumor-restricted virus infection and cell lysis. Clinical trials have shown that oncolytic adenoviruses are well tolerated in patients but also that their antitumor activity needs improvement. A promising strategy toward this end is to trigger systemic and prolonged antitumor immunity by adenoviral oncolysis. Antitumor immune activation depends in large part on antigen presentation and T cell activation by dendritic cells (DCs). Thus, it is likely that the interaction of lysed tumor cells with DCs is a key determinant of such "oncolytic vaccination." Our study reveals that human DCs effectively phagocytose melanoma cells at late stages of oncolytic adenovirus infection, when the cells die showing preferentially features of necrotic cell death. Maturation, migration toward CCL19 and T cell stimulatory capacity of DCs, crucial steps for immune induction, were, however, not induced by phagocytosis of oncolysate, but could be triggered by a cytokine maturation cocktail. Therefore, oncolytic adenoviruses and adenoviral oncolysate did not block DC maturation, which is in contrast to reports for other oncolytic viruses. These results represent a rationale for inserting immunostimulatory genes into oncolytic adenovirus genomes to assure critical DC maturation. Indeed, we report here that adenoviral transduction of melanoma cells with CD40L during oncolysis triggers the maturation of human DCs with T cell stimulatory capacity similar to DCs matured by cytokines. We conclude that triggering and shaping DC-induced antitumor immunity by oncolytic adenoviruses "armed" with immunostimulatory genes holds promise for improving the therapeutic outcome of viral oncolysis in patients.
Collapse
Affiliation(s)
- Stephan Schierer
- Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
79
|
Activation of cytotoxic and regulatory functions of NK cells by Sindbis viral vectors. PLoS One 2011; 6:e20598. [PMID: 21674047 PMCID: PMC3107224 DOI: 10.1371/journal.pone.0020598] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2011] [Accepted: 05/05/2011] [Indexed: 01/19/2023] Open
Abstract
Oncolytic viruses (OVs) represent a relatively novel anti-cancer modality. Like other new cancer treatments, effective OV therapy will likely require combination with conventional treatments. In order to design combinatorial treatments that work well together, a greater scrutiny of the mechanisms behind the individual treatments is needed. Sindbis virus (SV) based vectors have previously been shown to target and kill tumors in xenograft, syngeneic, and spontaneous mouse models. However, the effect of SV treatment on the immune system has not yet been studied. Here we used a variety of methods, including FACS analysis, cytotoxicity assays, cell depletion, imaging of tumor growth, cytokine blockade, and survival experiments, to study how SV therapy affects Natural Killer (NK) cell function in SCID mice bearing human ovarian carcinoma tumors. Surprisingly, we found that SV anti-cancer efficacy is largely NK cell-dependent. Furthermore, the enhanced therapeutic effect previously observed from Sin/IL12 vectors, which carry the gene for interleukin 12, is also NK cell dependent, but works through a separate IFNγ-dependent mechanism, which also induces the activation of peritoneal macrophages. These results demonstrate the multimodular nature of SV therapy, and open up new possibilities for potential synergistic or additive combinatorial therapies with other treatments.
Collapse
|
80
|
Melcher A, Parato K, Rooney CM, Bell JC. Thunder and lightning: immunotherapy and oncolytic viruses collide. Mol Ther 2011; 19:1008-16. [PMID: 21505424 DOI: 10.1038/mt.2011.65] [Citation(s) in RCA: 178] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
For the last several decades, the development of antitumor immune-based strategies and the engineering and testing of oncolytic viruses (OVs) has occurred largely in parallel tracks. Indeed, the immune system is often thought of as an impediment to successful oncolytic virus delivery and efficacy. More recently, however, both preclinical and clinical results have revealed potential synergy between these two promising therapeutic strategies. Here, we summarize some of the evidence that supports combining OVs with immuno-therapeutics and suggest new ways to mount a multipronged biological attack against cancers.
Collapse
Affiliation(s)
- Alan Melcher
- Targeted and Biological Therapies Group, Leeds Institute of Molecular Medicine, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, UK
| | | | | | | |
Collapse
|
81
|
Abstract
Oncolytic virotherapy is an emerging experimental treatment platform for cancer therapy. Oncolytic viruses are replicative-competent viruses that are engineered to replicate selectively in cancer cells with specified oncogenic phenotypes. Multiple DNA and RNA viruses have been clinically tested in a variety of tumors. This review will provide a brief description of these novel anticancer biologics and will summarize the results of clinical investigation. To date oncolytic virotherapy has shown to be safe, and has generated clinical responses in tumors that are resistant to chemotherapy or radiotherapy. The major challenge for researchers is to maximize the efficacy of these viral therapeutics, and to establish stable systemic delivery mechanisms.
Collapse
|
82
|
Horst D, Ressing ME, Wiertz EJHJ. Exploiting human herpesvirus immune evasion for therapeutic gain: potential and pitfalls. Immunol Cell Biol 2011; 89:359-66. [PMID: 21301483 DOI: 10.1038/icb.2010.129] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Herpesviruses stand out for their capacity to establish lifelong infections of immunocompetent hosts, generally without causing overt symptoms. Herpesviruses are equipped with sophisticated immune evasion strategies, allowing these viruses to persist for life despite the presence of a strong antiviral immune response. Although viral evasion tactics appear to target virtually any stage of the innate and adaptive host immune response, detailed knowledge is now available on the molecular mechanisms underlying herpesvirus obstruction of MHC class I-restricted antigen presentation to T cells. This opens the way for clinical application. Here, we review and discuss recent efforts to exploit human herpesvirus MHC class I evasion strategies for the rational design of novel strategies for vaccine development, cancer treatment, transplant protection and gene therapy.
Collapse
Affiliation(s)
- Daniëlle Horst
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | |
Collapse
|
83
|
Oncolytic virotherapy of gynecologic malignancies. Gynecol Oncol 2011; 120:302-10. [DOI: 10.1016/j.ygyno.2010.10.031] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Revised: 10/21/2010] [Accepted: 10/24/2010] [Indexed: 11/20/2022]
|
84
|
Geevarghese SK, Geller DA, de Haan HA, Hörer M, Knoll AE, Mescheder A, Nemunaitis J, Reid TR, Sze DY, Tanabe KK, Tawfik H. Phase I/II study of oncolytic herpes simplex virus NV1020 in patients with extensively pretreated refractory colorectal cancer metastatic to the liver. Hum Gene Ther 2010; 21:1119-28. [PMID: 20486770 DOI: 10.1089/hum.2010.020] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
This multicenter phase I/II study evaluated the safety, pharmacokinetics, and antitumor effects of repeated doses of NV1020, a genetically engineered oncolytic herpes simplex virus, in patients with advanced metastatic colorectal cancer (mCRC). Patients with liver-dominant mCRC received four fixed NV1020 doses via weekly hepatic artery infusion, followed by two or more cycles of conventional chemotherapy. Phase I included cohorts receiving 3 × 10(6), 1 × 10(7), 3 × 10(7), and 1 × 10(8) plaque-forming units (PFU)/dose to determine the optimal biological dose (OBD) for phase II. Blind independent computed tomography scan review was based on RECIST (response evaluation criteria in solid tumors) to assess hepatic tumor response. Phase I and II enrolled 13 and 19 patients, respectively. Patients experienced transient mild-moderate febrile reactions after each NV1020 infusion. Grade 3/4 virus-related toxicity was limited to transient lymphopenia in two patients. NV1020 shedding was not detected. Simultaneous cytokine and grade 1 coagulation perturbations were dose-limiting at 1 × 10(8) PFU/dose, considered the OBD. All 22 OBD patients had previously received 5-fluorouracil; most had received oxaliplatin or irinotecan (50% had both), many with at least one targeted agent. After NV1020 administration, 50% showed stable disease. The best overall tumor control rate after chemotherapy was 68% (1 partial response, 14 stable disease); this did not correlate with baseline variables or chemotherapy. Median time to progression was 6.4 months (95% confidence interval: 2, 8.9); median overall survival was 11.8 months (95% confidence interval: 8.3, 20.7). One-year survival was 47.2%. We conclude that NV1020 stabilizes liver metastases with minimal toxicity in mCRC. It may resensitize metastases to salvage chemotherapy and extend overall survival. A randomized phase II/III trial now appears justified.
Collapse
Affiliation(s)
- Sunil K Geevarghese
- Department of Hepatobiliary Surgery and Liver Transplantation, Vanderbilt University, Nashville, TN 37232, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Hammill AM, Conner J, Cripe TP. Oncolytic virotherapy reaches adolescence. Pediatr Blood Cancer 2010; 55:1253-63. [PMID: 20734404 DOI: 10.1002/pbc.22724] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2010] [Accepted: 06/01/2010] [Indexed: 01/11/2023]
Abstract
Lytic viruses kill cells as a consequence of their normal replication life cycle. The idea of harnessing viruses to kill cancer cells arose over a century ago, before viruses were even discovered, from medical case reports of infections associated with cancer remissions. Since then, there has been no shortage of hype, hope, or fear regarding the prospect of oncolytic virotherapy for cancer. Early developments in the field included encouraging antitumor efficacy both in animal studies in the 1920s-1940s and in human clinical trials in the 1950s-1970s. Despite its long-standing history, oncolytic virotherapy was an idea ahead of its time. Without needed advances in molecular biology, virology, immunology, and clinical research ethics, early clinical trials resulted in infectious complications and were fraught with controversial research conduct, so that enthusiasm in the medical community waned. Oncolytic virotherapy is now experiencing a major growth spurt, having sustained numerous laboratory advances and undergone multiple encouraging adult clinical trials, and is now witnessing the emergence of pediatric trials. Here we review the history and salient biology of the field, including preclinical and clinical data, with a special emphasis on those agents now being tested in pediatric cancer patients.
Collapse
Affiliation(s)
- Adrienne M Hammill
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | | |
Collapse
|
86
|
Sobol PT, Boudreau JE, Stephenson K, Wan Y, Lichty BD, Mossman KL. Adaptive antiviral immunity is a determinant of the therapeutic success of oncolytic virotherapy. Mol Ther 2010; 19:335-44. [PMID: 21119618 DOI: 10.1038/mt.2010.264] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Oncolytic virotherapy, the selective killing of tumor cells by oncolytic viruses (OVs), has emerged as a promising avenue of anticancer research. We have previously shown that KM100, a Herpes simplex virus type-1 (HSV) deficient for infected cell protein 0 (ICP0), possesses substantial oncolytic properties in vitro and has antitumor efficacy in vivo, in part by inducing antitumor immunity. Here, we illustrate through T-cell immunodepletion studies in nontolerized tumor-associated antigen models of breast cancer that KM100 treatment promotes antiviral and antitumor CD8(+) cytotoxic T-cell responses necessary for complete tumor regression. In tolerized tumor-associated antigen models of breast cancer, antiviral CD8(+) cytotoxic T-cell responses against infected tumor cells correlated with the induction of significant tumoristasis in the absence of tumor-associated antigen-specific CD8(+) cytotoxic T-cells. To enhance oncolysis, we tested a more cytopathic ICP0-null HSV and a vesicular stomatitis virus M protein mutant and found that despite improved in vitro replication, oncolysis in vivo did not improve. These studies illustrate that the in vitro cytolytic properties of OVs are poor prognostic indicators of in vivo antitumor activity, and underscore the importance of adaptive antiviral CD8(+) cytotoxic T-cells in effective cancer virotherapy.
Collapse
Affiliation(s)
- Paul T Sobol
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
87
|
Kanzaki M, Okamoto T, Mitsui H, Shibagaki N, Shimada S. A novel immunotherapeutic approach to melanoma-bearing hosts with protein-transduction domain-containing immunogenic foreign antigens. J Dermatol Sci 2010; 60:84-94. [DOI: 10.1016/j.jdermsci.2010.08.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Revised: 07/28/2010] [Accepted: 08/19/2010] [Indexed: 01/15/2023]
|
88
|
Willmon C, Diaz RM, Wongthida P, Galivo F, Kottke T, Thompson J, Albelda S, Harrington K, Melcher A, Vile R. Vesicular stomatitis virus-induced immune suppressor cells generate antagonism between intratumoral oncolytic virus and cyclophosphamide. Mol Ther 2010; 19:140-9. [PMID: 20978474 DOI: 10.1038/mt.2010.224] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Despite having potent oncolytic activity, in vitro, direct intratumoral injection of oncolytic vesicular stomatitis virus (VSV) into established AE17ova mesothelioma tumors in C57Bl/6 mice had no therapeutic effect. During studies to combine systemic cyclophosphamide (CPA) with VSV to suppress the innate immune reaction against VSV, we observed that CPA alone had highly significant antitumor effects in this model. However, against our expectations, the combination of CPA and VSV consistently reduced therapeutic efficacy compared to CPA alone, despite the fact that the combination increased intratumoral VSV titers. We show here that CPA-mediated therapy against AE17ova tumors was immune-mediated and dependent upon both CD4 T cells and natural killer (NK) cells. However, intratumoral VSV induced a transforming growth factor-β (TGF-β)-dependent suppressive activity, mediated by CD11b(+)GR-1(+) cells that significantly inhibited both antigen-specific T-cell activation, and CPA-activated, NK-dependent killing of AE17ova tumor cells. Overall, our results show that treatment with oncolytic viruses can induce a variety of immune-mediated consequences in vivo with both positive, or negative, effects on antitumor therapy. These underexplored immune consequences of treatment with oncolytic viruses may have significant, and possibly unexpected, impacts on how virotherapy interacts in combination with other agents which modulate antitumor immune effectors.
Collapse
Affiliation(s)
- Candice Willmon
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Wongthida P, Diaz RM, Galivo F, Kottke T, Thompson J, Melcher A, Vile R. VSV oncolytic virotherapy in the B16 model depends upon intact MyD88 signaling. Mol Ther 2010; 19:150-8. [PMID: 20959810 DOI: 10.1038/mt.2010.225] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We show here, for the first time to our knowledge, that the antitumor therapy of oncolytic vesicular stomatitis virus (VSV) in the B16ova model depends upon signaling through myeloid differentiation primary response gene 88 (MyD88) in host cells. VSV-mediated therapy of B16ova tumors was abolished in MyD88(-/-) mice despite generation of antigen-specific T cell responses similar to those in immune-competent mice. Mice defective in only toll-like receptor 4 (TLR4), TLR7, or interleukin 1 (IL-1) signaling retained VSV-induced therapy, suggesting that multiple, redundant pathways of innate immune activation by the virus contribute to antitumor immune reactivity. Lack of MyD88 signaling was associated with decreased expression of proinflammatory cytokines and neutrophil infiltration in response to intratumoral virus, as well as decreased infiltration of draining lymph nodes (LN) with plasmacytoid dendritic cells (pDCs) (CD11b(-)GR1(+)B220(+)) and myeloid-derived suppressor cells (CD11b(+)GR1(+)F4/80(+)). MyD88 signaling in response to VSV was also closely associated with a type I interferon (IFN) response. This inhibited virus replication within the tumor but also protected the host from viral dissemination from the tumor. Therefore, the innate immune response to oncolytic viruses can be, simultaneously, protherapeutic, antioncolytic, and systemically protective. These paradoxically conflicting roles need to be carefully considered in future strategies designed to improve the efficacy of oncolytic virotherapy.
Collapse
|
90
|
Kanai R, Wakimoto H, Cheema T, Rabkin SD. Oncolytic herpes simplex virus vectors and chemotherapy: are combinatorial strategies more effective for cancer? Future Oncol 2010; 6:619-34. [PMID: 20373873 DOI: 10.2217/fon.10.18] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Despite aggressive treatments, including chemotherapy and radiotherapy, cancers often recur owing to resistance to conventional therapies. Oncolytic viruses such as oncolytic herpes simplex virus (oHSV) represent an exciting biological approach to cancer therapy. A range of viral mutations has been engineered into HSV to engender oncolytic activity. While oHSV as a single agent has been tested in a number of cancer clinical trials, preclinical studies have demonstrated enhanced efficacy when it is combined with cytotoxic anticancer drugs. Among the strategies that will be discussed in this article are combinations with standard-of-care chemotherapeutics, expression of prodrug-activating enzymes to enhance chemotherapy and small-molecule inhibitors. The combination of oHSV and chemotherapy can achieve much more efficient cancer cell killing than either single agent alone, often through synergistic interactions. This can be clinically important not just for improving efficacy but also for permitting lower and less toxic chemotherapeutic doses. The viral mutations in an oHSV vector often determine the favorability of its interactions with chemotherapy, just as different cancer cells, due to genetic alterations, vary in their response to chemotherapy. As chemotherapeutics are often the standard of care, combining them with an investigational new drug, such as oHSV, is clinically easier than combining multiple novel agents. As has become clear for most cancer therapies, multimodal treatments are usually more effective. In this article, we will discuss the recent progress of these combinatorial strategies between virotherapy and chemotherapy and future directions.
Collapse
Affiliation(s)
- Ryuichi Kanai
- Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital, & Harvard Medical School, Boston, MA, USA
| | | | | | | |
Collapse
|
91
|
Marconi P, Argnani R, Epstein AL, Manservigi R. HSV as a vector in vaccine development and gene therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 655:118-44. [PMID: 20047039 DOI: 10.1007/978-1-4419-1132-2_10] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The very deep knowledge acquired on the genetics and molecular biology of herpes simplex virus (HSV), major human pathogen whose lifestyle is based on a long-term dual interaction with the infected host characterized by the existence of lytic and latent infections, has allowed the development of potential vectors for several applications in human healthcare. These include delivery and expression of human genes to cells of the nervous system, selective destruction of cancer cells, prophylaxis against infection with HSV or other infectious diseases and targeted infection of specific tissues or organs. Three different classes of vectors can be derived from HSV-1: replication-competent attenuated vectors, replication-incompetent recombinant vectors and defective helper-dependent vectors known as amplicons. This chapter highlights the current knowledge concerning design, construction and recent applications, as well as the potential and current limitations of the three different classes of HSV-1-based vectors.
Collapse
Affiliation(s)
- Peggy Marconi
- Department of Experimental and Diagnostic Medicine-Section of Microbiology, University of Ferrara, Via Luigi Borsari 46, Ferrara, 44100, Italy.
| | | | | | | |
Collapse
|
92
|
Abstract
The very deep knowledge acquired on the genetics and molecular biology of herpes simplex virus (HSV), has allowed the development of potential replication-competent and replication-defective vectors for several applications in human healthcare. These include delivery and expression of human genes to cells of the nervous systems, selective destruction of cancer cells, prophylaxis against infection with HSV or other infectious diseases, and targeted infection to specific tissues or organs. Replication-defective recombinant vectors are non-toxic gene transfer tools that preserve most of the neurotropic features of wild type HSV-1, particularly the ability to express genes after having established latent infections, and are thus proficient candidates for therapeutic gene transfer settings in neurons. A replication-defective HSV vector for the treatment of pain has recently entered in phase 1 clinical trial. Replication-competent (oncolytic) vectors are becoming a suitable and powerful tool to eradicate brain tumours due to their ability to replicate and spread only within the tumour mass, and have reached phase II/III clinical trials in some cases. The progress in understanding the host immune response induced by the vector is also improving the use of HSV as a vaccine vector against both HSV infection and other pathogens. This review briefly summarizes the obstacle encountered in the delivery of HSV vectors and examines the various strategies developed or proposed to overcome such challenges.
Collapse
Affiliation(s)
- Roberto Manservigi
- Department of Experimental and Diagnostic Medicine - Section of Microbiology, University of Ferrara, Via Luigi Borsari 46, 44100 Ferrara, Italy
| | | | | |
Collapse
|
93
|
Manservigi R, Argnani R, Marconi P. HSV Recombinant Vectors for Gene Therapy. Open Virol J 2010; 4:123-56. [PMID: 20835362 DOI: 10.2174/1874357901004030123] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Revised: 03/13/2010] [Accepted: 03/31/2010] [Indexed: 12/16/2022] Open
Abstract
The very deep knowledge acquired on the genetics and molecular biology of herpes simplex virus (HSV), has allowed the development of potential replication-competent and replication-defective vectors for several applications in human healthcare. These include delivery and expression of human genes to cells of the nervous systems, selective destruction of cancer cells, prophylaxis against infection with HSV or other infectious diseases, and targeted infection to specific tissues or organs. Replication-defective recombinant vectors are non-toxic gene transfer tools that preserve most of the neurotropic features of wild type HSV-1, particularly the ability to express genes after having established latent infections, and are thus proficient candidates for therapeutic gene transfer settings in neurons. A replication-defective HSV vector for the treatment of pain has recently entered in phase 1 clinical trial. Replication-competent (oncolytic) vectors are becoming a suitable and powerful tool to eradicate brain tumours due to their ability to replicate and spread only within the tumour mass, and have reached phase II/III clinical trials in some cases. The progress in understanding the host immune response induced by the vector is also improving the use of HSV as a vaccine vector against both HSV infection and other pathogens. This review briefly summarizes the obstacle encountered in the delivery of HSV vectors and examines the various strategies developed or proposed to overcome such challenges.
Collapse
Affiliation(s)
- Roberto Manservigi
- Department of Experimental and Diagnostic Medicine - Section of Microbiology, University of Ferrara, Via Luigi Borsari 46, 44100 Ferrara, Italy
| | | | | |
Collapse
|
94
|
Bridle BW, Stephenson KB, Boudreau JE, Koshy S, Kazdhan N, Pullenayegum E, Brunellière J, Bramson JL, Lichty BD, Wan Y. Potentiating cancer immunotherapy using an oncolytic virus. Mol Ther 2010; 18:1430-9. [PMID: 20551919 DOI: 10.1038/mt.2010.98] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Oncolytic viruses (OVs) are highly immunogenic and this limits their use in immune-competent hosts. Although immunosuppression may improve viral oncolysis, this gain is likely achieved at the cost of antitumoral immunity. We have developed a strategy wherein the immune response against the OV leads to enhanced therapeutic outcomes. We demonstrate that immunization with an adenoviral (Ad) vaccine before treatment with an oncolytic vesicular stomatitis virus (VSV) expressing the same tumor antigen (Ag) leads to significantly enhanced antitumoral immunity. Intratumoral replication of VSV was minimally attenuated in Ad-immunized hosts but extending the interval between treatments reduced the attenuating effect and further increased antitumoral immunity. More importantly, our combination approach shifted the immune response from viral Ags to tumor Ags and further reduced OV replication in normal tissues, leading to enhancements in both efficacy and safety. These studies also highlight the benefits of using a replicating, OV to boost a pre-existing antitumoral immune response as this approach generated larger responses versus tumor Ag in tumor-bearing hosts than could be achieved in tumor-free hosts. This strategy should be applicable to other vector combinations, tumor Ags, and tumor targets.
Collapse
Affiliation(s)
- Byram W Bridle
- Centre for Gene Therapeutics, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Wongthida P, Diaz RM, Galivo F, Kottke T, Thompson J, Pulido J, Pavelko K, Pease L, Melcher A, Vile R. Type III IFN interleukin-28 mediates the antitumor efficacy of oncolytic virus VSV in immune-competent mouse models of cancer. Cancer Res 2010; 70:4539-49. [PMID: 20484025 DOI: 10.1158/0008-5472.can-09-4658] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Innate immune effector mechanisms triggered by oncolytic viruses may contribute to the clearance of both infected and uninfected tumor cells in immunocompetent murine hosts. Here, we developed an in vitro tumor cell/bone marrow coculture assay and used it to dissect innate immune sensor and effector responses to intratumoral vesicular stomatitis virus (VSV). We found that the type III IFN interleukin-28 (IL-28) was induced by viral activation of innate immune-sensing cells, acting as a key mediator of VSV-mediated virotherapy of B16ova melanomas. Using tumor variants which differentially express the IL-28 receptor, we showed that IL-28 induced by VSV within the tumor microenvironment sensitizes tumor cells to natural killer cell recognition and activation. These results revealed new insights into the immunovirological mechanisms associated with oncolytic virotherapy in immune-competent hosts. Moreover, they defined a new class of tumor-associated mutation, such as acquired loss of responsiveness to IL-28 signaling, which confers insensitivity to oncolytic virotherapy through a mechanism independent of viral replication in vitro. Lastly, the findings suggested new strategies to manipulate immune signals that may enhance viral replication, along with antitumor immune activation, and improve the efficacy of oncolytic virotherapies.
Collapse
MESH Headings
- Animals
- Bone Marrow Cells/immunology
- Bone Marrow Cells/virology
- Cytokines/biosynthesis
- Cytokines/immunology
- Cytotoxicity, Immunologic
- Disease Models, Animal
- Immunity, Innate/immunology
- Immunocompromised Host
- Injections, Intralesional
- Interferon Type I/immunology
- Killer Cells, Natural/immunology
- Melanoma, Experimental/immunology
- Melanoma, Experimental/therapy
- Melanoma, Experimental/virology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Oncolytic Virotherapy/methods
- Receptors, Cytokine/biosynthesis
- Receptors, Cytokine/immunology
- Vesicular stomatitis Indiana virus/immunology
- Virus Replication/immunology
Collapse
Affiliation(s)
- Phonphimon Wongthida
- Departments of Molecular Medicine, Immunology, and Ophthalmology and Ocular Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Castelo-Branco P, Passer BJ, Buhrman JS, Antoszczyk S, Marinelli M, Zaupa C, Rabkin SD, Martuza RL. Oncolytic herpes simplex virus armed with xenogeneic homologue of prostatic acid phosphatase enhances antitumor efficacy in prostate cancer. Gene Ther 2010; 17:805-10. [PMID: 20220784 DOI: 10.1038/gt.2010.20] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Prostate cancer is one of the most prevalent cancers in men. Replication-competent oncolytic herpes simplex virus (oHSV) vectors are a powerful antitumor therapy that can exert at least two effects: direct cytocidal activity that selectively kills cancer cells and induction of antitumor immunity. In addition, oHSV vectors can also function as a platform to deliver transgenes of interest. In these studies, we have examined the expression of a xenogeneic homologue of the prostate cancer antigen, prostatic acid phosphatase (PAP), with the goal of enhancing virotherapy against PAP-expressing tumors. PAP has already been used for cancer vaccination in patients with prostate cancer. Here we show that treatment with oHSV bPDelta6 expressing xenogeneic human PAP (hPAP) significantly reduces tumor growth and increases survival of C57/BL6 mice bearing mouse TRAMP-C2 prostate tumors, whereas expression of syngeneic mouse PAP (mPAP) from the same oHSV vector did not enhance antitumor activity. Treatment of mice bearing metastatic TRAMP-C2 lung tumors with oHSV-expressing hPAP resulted in fewer tumor nodules. To our knowledge, this is the first report of oncolytic viruses being used to express xenoantigens. These data lend support to the concept of combining oncolytic and immunogenic therapies as a way to improve therapy of metastatic prostate cancer.
Collapse
Affiliation(s)
- P Castelo-Branco
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | | | | | | | |
Collapse
|
97
|
Agarwalla PK, Barnard ZR, Curry WT. Virally mediated immunotherapy for brain tumors. Neurosurg Clin N Am 2009; 21:167-79. [PMID: 19944975 DOI: 10.1016/j.nec.2009.08.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Brain tumors are a leading cause of mortality and morbidity in the United States. Malignant brain tumors occur in approximately 80,000 adults. Furthermore, the average 5-year survival rate for malignant brain tumors across all ages and races is approximately 30% and has remained relatively static over the past few decades, showing the need for continued research and progress in brain tumor therapy. Improved techniques in molecular biology have expanded understanding of tumor genetics and permitted viral engineering and the anticancer therapeutic use of viruses as directly cytotoxic agents and as gene vectors. Preclinical models have shown promising antitumor effects, and generation of clinical grade vectors is feasible. In parallel to these developments, better understanding of antitumor immunity has been accompanied by progress in cancer immunotherapy, the goal of which is to stimulate host rejection of a growing tumor. This article reviews the intersection between the use of viral therapy and immunotherapy in the treatment of malignant gliomas. Each approach shows great promise on its own and in combined or integrated forms.
Collapse
Affiliation(s)
- Pankaj K Agarwalla
- Brain Tumor Immunotherapy Laboratory, Department of Neurosurgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
98
|
Ottolino-Perry K, Diallo JS, Lichty BD, Bell JC, McCart JA. Intelligent design: combination therapy with oncolytic viruses. Mol Ther 2009; 18:251-63. [PMID: 20029399 DOI: 10.1038/mt.2009.283] [Citation(s) in RCA: 146] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Metastatic cancer remains an incurable disease in the majority of cases and thus novel treatment strategies such as oncolytic virotherapy are rapidly advancing toward clinical use. In order to be successful, it is likely that some type of combination therapy will be necessary to have a meaningful impact on this disease. Although it may be tempting to simply combine an oncolytic virus with the existing standard radiation or chemotherapeutics, the long-term goal of such treatments must be to have a rational, potentially synergistic combination strategy that can be safely and easily used in the clinical setting. The combination of oncolytic virotherapy with existing radiotherapy and chemotherapy modalities is reviewed along with novel biologic therapies including immunotherapies, in order to help investigators make intelligent decisions during the clinical development of these products.
Collapse
Affiliation(s)
- Kathryn Ottolino-Perry
- Division of Experimental Therapeutics, Toronto General Research Institute, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
99
|
Senzer NN, Kaufman HL, Amatruda T, Nemunaitis M, Reid T, Daniels G, Gonzalez R, Glaspy J, Whitman E, Harrington K, Goldsweig H, Marshall T, Love C, Coffin R, Nemunaitis JJ. Phase II Clinical Trial of a Granulocyte-Macrophage Colony-Stimulating Factor–Encoding, Second-Generation Oncolytic Herpesvirus in Patients With Unresectable Metastatic Melanoma. J Clin Oncol 2009; 27:5763-71. [DOI: 10.1200/jco.2009.24.3675] [Citation(s) in RCA: 472] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
PurposeTreatment options for metastatic melanoma are limited. We conducted this phase II trial to assess the efficacy of JS1/34.5-/47-/granulocyte-macrophage colony-stimulating factor (GM-CSF) in stages IIIc and IV disease.Patients and MethodsTreatment involved intratumoral injection of up to 4 mL of 106pfu/mL of JS1/34.5-/47-/GM-CSF followed 3 weeks later by up to 4 mL of 108pfu/mL every 2 weeks for up to 24 treatments. Clinical activity (by RECIST [Response Evaluation Criteria in Solid Tumors]), survival, and safety parameters were monitored.ResultsFifty patients (stages IIIc, n = 10; IVM1a, n = 16; IVM1b, n = 4; IVM1c, n = 20) received a median of six injection sets; 74% of patients had received one or more nonsurgical prior therapies for active disease, including dacarbazine/temozolomide or interleukin-2 (IL-2). Adverse effects were limited primarily to transient flu-like symptoms. The overall response rate by RECIST was 26% (complete response [CR], n = 8; partial response [PR], n = 5), and regression of both injected and distant (including visceral) lesions occurred. Ninety-two percent of the responses had been maintained for 7 to 31 months. Ten additional patients had stable disease (SD) for greater than 3 months, and two additional patients had surgical CR. On an extension protocol, two patients subsequently achieved CR by 24 months (one previously PR, one previously SD), and one achieved surgical CR (previously PR). Overall survival was 58% at 1 year and 52% at 24 months.ConclusionThe 26% response rate, with durability in both injected and uninjected lesions including visceral sites, together with the survival rates, are evidence of systemic effectiveness. This effectiveness, combined with a limited toxicity profile, warrants additional evaluation of JS1/34.5-/47-/GM-CSF in metastatic melanoma. A US Food and Drug Administration–approved phase III investigation is underway.
Collapse
Affiliation(s)
- Neil N. Senzer
- From the Mary Crowley Cancer Research Centers; Texas Oncology Physicians Association; and Baylor Sammons Cancer Center, Dallas, TX; Columbia University, Department of Surgery, New York, NY; Hubert H. Humphrey Cancer Center, Robbinsdale, MN; University of Colorado, Aurora, CO; University of California, San Diego Cancer Center, La Jolla; and University of California, Los Angeles, Los Angeles, CA; Mountainside Hospital, Montclair, NJ; Royal Marsden Hospital, London, United Kingdom; and BioVex, Woburn, MA
| | - Howard L. Kaufman
- From the Mary Crowley Cancer Research Centers; Texas Oncology Physicians Association; and Baylor Sammons Cancer Center, Dallas, TX; Columbia University, Department of Surgery, New York, NY; Hubert H. Humphrey Cancer Center, Robbinsdale, MN; University of Colorado, Aurora, CO; University of California, San Diego Cancer Center, La Jolla; and University of California, Los Angeles, Los Angeles, CA; Mountainside Hospital, Montclair, NJ; Royal Marsden Hospital, London, United Kingdom; and BioVex, Woburn, MA
| | - Thomas Amatruda
- From the Mary Crowley Cancer Research Centers; Texas Oncology Physicians Association; and Baylor Sammons Cancer Center, Dallas, TX; Columbia University, Department of Surgery, New York, NY; Hubert H. Humphrey Cancer Center, Robbinsdale, MN; University of Colorado, Aurora, CO; University of California, San Diego Cancer Center, La Jolla; and University of California, Los Angeles, Los Angeles, CA; Mountainside Hospital, Montclair, NJ; Royal Marsden Hospital, London, United Kingdom; and BioVex, Woburn, MA
| | - Mike Nemunaitis
- From the Mary Crowley Cancer Research Centers; Texas Oncology Physicians Association; and Baylor Sammons Cancer Center, Dallas, TX; Columbia University, Department of Surgery, New York, NY; Hubert H. Humphrey Cancer Center, Robbinsdale, MN; University of Colorado, Aurora, CO; University of California, San Diego Cancer Center, La Jolla; and University of California, Los Angeles, Los Angeles, CA; Mountainside Hospital, Montclair, NJ; Royal Marsden Hospital, London, United Kingdom; and BioVex, Woburn, MA
| | - Tony Reid
- From the Mary Crowley Cancer Research Centers; Texas Oncology Physicians Association; and Baylor Sammons Cancer Center, Dallas, TX; Columbia University, Department of Surgery, New York, NY; Hubert H. Humphrey Cancer Center, Robbinsdale, MN; University of Colorado, Aurora, CO; University of California, San Diego Cancer Center, La Jolla; and University of California, Los Angeles, Los Angeles, CA; Mountainside Hospital, Montclair, NJ; Royal Marsden Hospital, London, United Kingdom; and BioVex, Woburn, MA
| | - Gregory Daniels
- From the Mary Crowley Cancer Research Centers; Texas Oncology Physicians Association; and Baylor Sammons Cancer Center, Dallas, TX; Columbia University, Department of Surgery, New York, NY; Hubert H. Humphrey Cancer Center, Robbinsdale, MN; University of Colorado, Aurora, CO; University of California, San Diego Cancer Center, La Jolla; and University of California, Los Angeles, Los Angeles, CA; Mountainside Hospital, Montclair, NJ; Royal Marsden Hospital, London, United Kingdom; and BioVex, Woburn, MA
| | - Rene Gonzalez
- From the Mary Crowley Cancer Research Centers; Texas Oncology Physicians Association; and Baylor Sammons Cancer Center, Dallas, TX; Columbia University, Department of Surgery, New York, NY; Hubert H. Humphrey Cancer Center, Robbinsdale, MN; University of Colorado, Aurora, CO; University of California, San Diego Cancer Center, La Jolla; and University of California, Los Angeles, Los Angeles, CA; Mountainside Hospital, Montclair, NJ; Royal Marsden Hospital, London, United Kingdom; and BioVex, Woburn, MA
| | - John Glaspy
- From the Mary Crowley Cancer Research Centers; Texas Oncology Physicians Association; and Baylor Sammons Cancer Center, Dallas, TX; Columbia University, Department of Surgery, New York, NY; Hubert H. Humphrey Cancer Center, Robbinsdale, MN; University of Colorado, Aurora, CO; University of California, San Diego Cancer Center, La Jolla; and University of California, Los Angeles, Los Angeles, CA; Mountainside Hospital, Montclair, NJ; Royal Marsden Hospital, London, United Kingdom; and BioVex, Woburn, MA
| | - Eric Whitman
- From the Mary Crowley Cancer Research Centers; Texas Oncology Physicians Association; and Baylor Sammons Cancer Center, Dallas, TX; Columbia University, Department of Surgery, New York, NY; Hubert H. Humphrey Cancer Center, Robbinsdale, MN; University of Colorado, Aurora, CO; University of California, San Diego Cancer Center, La Jolla; and University of California, Los Angeles, Los Angeles, CA; Mountainside Hospital, Montclair, NJ; Royal Marsden Hospital, London, United Kingdom; and BioVex, Woburn, MA
| | - Kevin Harrington
- From the Mary Crowley Cancer Research Centers; Texas Oncology Physicians Association; and Baylor Sammons Cancer Center, Dallas, TX; Columbia University, Department of Surgery, New York, NY; Hubert H. Humphrey Cancer Center, Robbinsdale, MN; University of Colorado, Aurora, CO; University of California, San Diego Cancer Center, La Jolla; and University of California, Los Angeles, Los Angeles, CA; Mountainside Hospital, Montclair, NJ; Royal Marsden Hospital, London, United Kingdom; and BioVex, Woburn, MA
| | - Howard Goldsweig
- From the Mary Crowley Cancer Research Centers; Texas Oncology Physicians Association; and Baylor Sammons Cancer Center, Dallas, TX; Columbia University, Department of Surgery, New York, NY; Hubert H. Humphrey Cancer Center, Robbinsdale, MN; University of Colorado, Aurora, CO; University of California, San Diego Cancer Center, La Jolla; and University of California, Los Angeles, Los Angeles, CA; Mountainside Hospital, Montclair, NJ; Royal Marsden Hospital, London, United Kingdom; and BioVex, Woburn, MA
| | - Tracey Marshall
- From the Mary Crowley Cancer Research Centers; Texas Oncology Physicians Association; and Baylor Sammons Cancer Center, Dallas, TX; Columbia University, Department of Surgery, New York, NY; Hubert H. Humphrey Cancer Center, Robbinsdale, MN; University of Colorado, Aurora, CO; University of California, San Diego Cancer Center, La Jolla; and University of California, Los Angeles, Los Angeles, CA; Mountainside Hospital, Montclair, NJ; Royal Marsden Hospital, London, United Kingdom; and BioVex, Woburn, MA
| | - Colin Love
- From the Mary Crowley Cancer Research Centers; Texas Oncology Physicians Association; and Baylor Sammons Cancer Center, Dallas, TX; Columbia University, Department of Surgery, New York, NY; Hubert H. Humphrey Cancer Center, Robbinsdale, MN; University of Colorado, Aurora, CO; University of California, San Diego Cancer Center, La Jolla; and University of California, Los Angeles, Los Angeles, CA; Mountainside Hospital, Montclair, NJ; Royal Marsden Hospital, London, United Kingdom; and BioVex, Woburn, MA
| | - Robert Coffin
- From the Mary Crowley Cancer Research Centers; Texas Oncology Physicians Association; and Baylor Sammons Cancer Center, Dallas, TX; Columbia University, Department of Surgery, New York, NY; Hubert H. Humphrey Cancer Center, Robbinsdale, MN; University of Colorado, Aurora, CO; University of California, San Diego Cancer Center, La Jolla; and University of California, Los Angeles, Los Angeles, CA; Mountainside Hospital, Montclair, NJ; Royal Marsden Hospital, London, United Kingdom; and BioVex, Woburn, MA
| | - John J. Nemunaitis
- From the Mary Crowley Cancer Research Centers; Texas Oncology Physicians Association; and Baylor Sammons Cancer Center, Dallas, TX; Columbia University, Department of Surgery, New York, NY; Hubert H. Humphrey Cancer Center, Robbinsdale, MN; University of Colorado, Aurora, CO; University of California, San Diego Cancer Center, La Jolla; and University of California, Los Angeles, Los Angeles, CA; Mountainside Hospital, Montclair, NJ; Royal Marsden Hospital, London, United Kingdom; and BioVex, Woburn, MA
| |
Collapse
|
100
|
Identification of virus resistant tumor cell subpopulations in three-dimensional uveal melanoma cultures. Cancer Gene Ther 2009; 17:223-34. [DOI: 10.1038/cgt.2009.73] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|