1
|
Novel Oncolytic Virus Armed with Cancer Suicide Gene and Normal Vasculogenic Gene for Improved Anti-Tumor Activity. Cancers (Basel) 2020; 12:cancers12051070. [PMID: 32344903 PMCID: PMC7281019 DOI: 10.3390/cancers12051070] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 04/24/2020] [Indexed: 12/22/2022] Open
Abstract
Here, we developed a novel oncolytic vaccinia virus (NOV) with the dual advantages of cancer selectivity and normal vessel reconstructive activity by replacing the viral thymidine kinase (vTk) and vaccinia growth factor (VGF) genes with genes encoding TNF-related apoptosis-inducing ligand (TRAIL) and angiopoietin 1 (Ang1), respectively. The pan-cancer-specific oncolytic potency of NOV was confirmed in various human and mouse cancer cell lines (colon, liver, pancreas, cholangiocarcinoma, cervical cancer, osteosarcoma, and melanoma). Vaccinia virus (VV) treatment directly induced early apoptosis in tumors within 24 h, and this effect was enhanced with further engineering; VGF and Tk deletion with Ang1 and TRAIL insertion. Meanwhile, treatment with the conventional anti-cancer drug cisplatin did not induce apoptosis. A virus-treated CT26 mouse colon cancer syngeneic model showed attenuated tumor growth, which was in accordance with the results of percent survival measurement, CD8 expression analysis, and TUNEL staining with advanced genetic engineering (vAng1 < vTRAIL < NOV). Taken together, our results indicate that NOV induces cancer tissue apoptosis and anti-tumor immunity and may constitute a highly advantageous therapeutic agent for next-generation solid tumor virotherapy with pan-cancer-specific oncolytic activity and high biosafety.
Collapse
|
2
|
Sobhanimonfared F, Bamdad T, Sadigh ZA, Choobin H. Virus specific tolerance enhanced efficacy of cancer immuno-virotherapy. Microb Pathog 2019; 140:103957. [PMID: 31891795 DOI: 10.1016/j.micpath.2019.103957] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 12/22/2019] [Accepted: 12/27/2019] [Indexed: 01/14/2023]
Abstract
BACKGROUND Activation of the immune system to fight cancer is a major goal in immunology and oncology. Although cancer treatment using oncolytic viruses shows promising results, virus mediated oncolysis induces a weak anti-tumor immune response. Upon application of viruses, immune responses against the virus play a significant role in limiting tumor virotherapy. Although suppression of host immunity increases the efficacy of virotherapy against the tumor, but inhibits anti-tumor immune responses. Induction of viral specific tolerance before viral replication may cause the virus to efficiently replicate in tumor cells without affecting the immune responses against tumor antigens. Investigation of the combined strategy of virotherapy and immunotherapy using irradiated tumor cells along with IL-2 and interferon-alpha in virus specific tolerant mice was the goal of this study. MATERIALS AND METHODS For tolerance induction, the newborn mice were injected with vesicular stomatitis virus (VSV) subcutaneously. After injection of TC-1 tumor cells to adult tolerant mice and formation of a tumor, irradiated TC-1 cells along with IL-2 and Interferon-alpha expression plasmid were injected twice in mice and followed by virotherapy. Size of tumors and CTL activity against the virus and tumor cells were measured. RESULT The results showed increased efficacy of virotherapy in combination with immune-stimulators and tumor cells injection in tolerant mice compared to normal mice. CONCLUSION Specific tolerance against the oncolytic virus enhances the efficacy of virotherapy both in monotherapy and in combination with immunotherapy.
Collapse
Affiliation(s)
- Fatemeh Sobhanimonfared
- Department of Virology, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Taravat Bamdad
- Department of Virology, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Zohreh Azita Sadigh
- Human Viral Vaccine Department, Razi Vaccine and Serum Research Institute, Karaj, Iran.
| | - Hamzeh Choobin
- Department of Virology, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
3
|
Zonov E, Kochneva G, Yunusova A, Grazhdantseva A, Richter V, Ryabchikova E. Features of the Antitumor Effect of Vaccinia Virus Lister Strain. Viruses 2016; 8:E20. [PMID: 26771631 PMCID: PMC4728580 DOI: 10.3390/v8010020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 12/30/2015] [Accepted: 01/06/2016] [Indexed: 02/06/2023] Open
Abstract
Oncolytic abilities of vaccinia virus (VACV) served as a basis for the development of various recombinants for treating cancer; however, "natural" oncolytic properties of the virus are not examined in detail. Our study was conducted to know how the genetically unmodified L-IVP strain of VACV produces its antitumor effect. Human A431 carcinoma xenografts in nude mice and murine Ehrlich carcinoma in C57Bl mice were used as targets for VACV, which was injected intratumorally. A set of virological methods, immunohistochemistry, light and electron microscopy was used in the study. We found that in mice bearing A431 carcinoma, the L-IVP strain was observed in visceral organs within two weeks, but rapidly disappeared from the blood. The L-IVP strain caused decrease of sizes in both tumors, however, in different ways. Direct cell destruction by replicating virus plays a main role in regression of A431 carcinoma xenografts, while in Ehrlich carcinoma, which poorly supported VACV replication, the virus induced decrease of mitoses by pushing tumor cells into S-phase of cell cycle. Our study showed that genetically unmodified VACV possesses at least two mechanisms of antitumor effect: direct destruction of tumor cells and suppression of mitoses in tumor cells.
Collapse
Affiliation(s)
- Evgeniy Zonov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences (ICBFM SB RAS), 8 Lavrentiev Avenue, Novosibirsk 630090, Russia.
| | - Galina Kochneva
- State Research Center of Virology and Biotechnology "Vector", Koltsovo 630559, Russia.
| | - Anastasiya Yunusova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences (ICBFM SB RAS), 8 Lavrentiev Avenue, Novosibirsk 630090, Russia.
| | | | - Vladimir Richter
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences (ICBFM SB RAS), 8 Lavrentiev Avenue, Novosibirsk 630090, Russia.
| | - Elena Ryabchikova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences (ICBFM SB RAS), 8 Lavrentiev Avenue, Novosibirsk 630090, Russia.
| |
Collapse
|
4
|
Jakeman PG, Hills TE, Tedcastle AB, Di Y, Fisher KD, Seymour LW. Improved in vitro human tumor models for cancer gene therapy. Hum Gene Ther 2015; 26:249-56. [PMID: 25808057 DOI: 10.1089/hum.2015.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Developing effective anticancer treatments is a particular challenge, as agents must contend with not only the target cellular biology, but also with the complex tumor microenvironment. Here we discuss various in vitro strategies that have sought to address this issue, with a particular focus on new methodologies that utilize clinical samples in basic research and their application in gene therapy and virotherapy.
Collapse
Affiliation(s)
- Philip G Jakeman
- Department of Oncology, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - Thomas E Hills
- Department of Oncology, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - Alison B Tedcastle
- Department of Oncology, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - Ying Di
- Department of Oncology, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - Kerry D Fisher
- Department of Oncology, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - Leonard W Seymour
- Department of Oncology, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| |
Collapse
|
5
|
Weichert JP, Clark PA, Kandela IK, Vaccaro AM, Clarke W, Longino MA, Pinchuk AN, Farhoud M, Swanson KI, Floberg JM, Grudzinski J, Titz B, Traynor AM, Chen HE, Hall LT, Pazoles CJ, Pickhardt PJ, Kuo JS. Alkylphosphocholine analogs for broad-spectrum cancer imaging and therapy. Sci Transl Med 2015; 6:240ra75. [PMID: 24920661 DOI: 10.1126/scitranslmed.3007646] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Many solid tumors contain an overabundance of phospholipid ethers relative to normal cells. Capitalizing on this difference, we created cancer-targeted alkylphosphocholine (APC) analogs through structure-activity analyses. Depending on the iodine isotope used, radioiodinated APC analog CLR1404 was used as either a positron emission tomography (PET) imaging ((124)I) or molecular radiotherapeutic ((131)I) agent. CLR1404 analogs displayed prolonged tumor-selective retention in 55 in vivo rodent and human cancer and cancer stem cell models. (131)I-CLR1404 also displayed efficacy (tumor growth suppression and survival extension) in a wide range of human tumor xenograft models. Human PET/CT (computed tomography) and SPECT (single-photon emission computed tomography)/CT imaging in advanced-cancer patients with (124)I-CLR1404 or (131)I-CLR1404, respectively, demonstrated selective uptake and prolonged retention in both primary and metastatic malignant tumors. Combined application of these chemically identical APC-based radioisosteres will enable personalized dual modality cancer therapy of using molecular (124)I-CLR1404 tumor imaging for planning (131)I-CLR1404 therapy.
Collapse
Affiliation(s)
- Jamey P Weichert
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA. Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA. Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA. Cellectar Biosciences Inc., 3301 Agriculture Drive, Madison, WI 53716, USA.
| | - Paul A Clark
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Irawati K Kandela
- Cellectar Biosciences Inc., 3301 Agriculture Drive, Madison, WI 53716, USA
| | - Abram M Vaccaro
- Cellectar Biosciences Inc., 3301 Agriculture Drive, Madison, WI 53716, USA
| | - William Clarke
- Cellectar Biosciences Inc., 3301 Agriculture Drive, Madison, WI 53716, USA
| | - Marc A Longino
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA. Cellectar Biosciences Inc., 3301 Agriculture Drive, Madison, WI 53716, USA
| | - Anatoly N Pinchuk
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA. Cellectar Biosciences Inc., 3301 Agriculture Drive, Madison, WI 53716, USA
| | - Mohammed Farhoud
- Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Kyle I Swanson
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - John M Floberg
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Joseph Grudzinski
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA. Cellectar Biosciences Inc., 3301 Agriculture Drive, Madison, WI 53716, USA
| | - Benjamin Titz
- Cellectar Biosciences Inc., 3301 Agriculture Drive, Madison, WI 53716, USA
| | - Anne M Traynor
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Hong-En Chen
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Lance T Hall
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | | | - Perry J Pickhardt
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - John S Kuo
- Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA. Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA.
| |
Collapse
|
6
|
Zamarin D, Wolchok JD. Potentiation of immunomodulatory antibody therapy with oncolytic viruses for treatment of cancer. MOLECULAR THERAPY-ONCOLYTICS 2014; 1:14004. [PMID: 27119094 PMCID: PMC4782939 DOI: 10.1038/mto.2014.4] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 06/30/2014] [Indexed: 02/07/2023]
Abstract
Identification of the immune suppressive mechanisms active within the tumor microenvironment led to development of immunotherapeutic strategies aiming to reverse the immunosuppression and to enhance the function of tumor-infiltrating lymphocytes. Of those, cancer therapy with antibodies targeting the immune costimulatory and coinhibitory receptors has demonstrated significant promise in the recent years, with multiple antibodies entering clinical testing. The responses to these agents, however, have not been universal and have not been observed in all cancer types, calling for identification of appropriate predictive biomarkers and development of combinatorial strategies. Pre-existing immune infiltration in tumors has been demonstrated to have a strong association with response to immunotherapies, with the type I interferon (IFN) pathway emerging as a key player in tumor innate immune recognition and activation of adaptive immunity. These findings provide a rationale for evaluation of strategies targeting the type I IFN pathway as a means to enhance tumor immune recognition and infiltration, which could potentially make them susceptible to therapeutics targeting the cosignaling receptors. To this end in particular, oncolytic viruses (OVs) have been demonstrated to enhance tumor recognition by the immune system through multiple mechanisms, which include upregulation of major histocompatibility complex and costimulatory molecules on cancer cells, immunogenic cell death and antigen release, and activation of the type I IFN pathway. Evidence is now emerging that combination therapies using OVs and agents targeting immune cosignaling receptors such as 4-1BB, PD-1, and CTLA-4 may work in concert to enhance antitumor immunity and therapeutic efficacy. Our evolving understanding of the interplay between OVs and the immune system demonstrates that the virus-induced antitumor immune responses can be harnessed to drive the efficacy of the agents targeting cosignaling receptors and provides a strong rationale for integration of such therapies in clinic.
Collapse
Affiliation(s)
- Dmitriy Zamarin
- Swim Across America Laboratory, Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, New York, USA; Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA; Ludwig Center for Cancer Immunotherapy at Memorial Sloan-Kettering Cancer Center, New York, New York, USA; Weill Cornell Medical College and Graduate School of Medical Sciences of Cornell University, New York, New York, USA
| | - Jedd D Wolchok
- Swim Across America Laboratory, Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, New York, USA; Department of Medicine, Melanoma and Immunotherapy Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA; Ludwig Center for Cancer Immunotherapy at Memorial Sloan-Kettering Cancer Center, New York, New York, USA; Weill Cornell Medical College and Graduate School of Medical Sciences of Cornell University, New York, New York, USA
| |
Collapse
|
7
|
Ultrasound-enhanced drug delivery in prostate cancer xenografts by nanoparticles stabilizing microbubbles. J Control Release 2014; 187:39-49. [DOI: 10.1016/j.jconrel.2014.05.020] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 05/07/2014] [Accepted: 05/09/2014] [Indexed: 11/20/2022]
|
8
|
Cronin M, Le Boeuf F, Murphy C, Roy DG, Falls T, Bell JC, Tangney M. Bacterial-mediated knockdown of tumor resistance to an oncolytic virus enhances therapy. Mol Ther 2014; 22:1188-1197. [PMID: 24569832 DOI: 10.1038/mt.2014.23] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 02/12/2014] [Indexed: 12/14/2022] Open
Abstract
Oncolytic viruses (OVs) and bacteria share the property of tumor-selective replication following systemic administration. In the case of nonpathogenic bacteria, tumor selectivity relates to their ability to grow extracellularly within tumor stroma and is therefore ideally suited to restricting the production of bacterially produced therapeutic agents to tumors. We have previously shown the ability of the type 1 interferon antagonist B18R to enhance the replication and spread of vesicular stomatitis virus (VSV) by overcoming related cellular innate immunity. In this study, we utilized nonpathogenic bacteria (E. coli) expressing B18R to facilitate tumor-specific production of B18R, resulting in a microenvironment depleted of bioactive antiviral cytokine, thus "preconditioning" the tumor to enhance subsequent tumor destruction by the OV. Both in vitro and in vivo infection by VSVΔ51 was greatly enhanced by B18R produced from E. coli. Moreover, a significant increase in therapeutic efficacy resulted from intravenous (i.v.) injection of bacteria to tumor-bearing mice 5 days prior to i.v. VSVΔ51 administration, as evidenced by a significant reduction in tumor growth and increased survival in mice. Our strategy is the first example where two such diverse microorganisms are rationally combined and demonstrates the feasibility of combining complementary microorganisms to improve therapeutic outcome.
Collapse
Affiliation(s)
- Michelle Cronin
- Cork Cancer Research Centre, University College Cork, Cork, Ireland
| | - Fabrice Le Boeuf
- Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Carola Murphy
- Cork Cancer Research Centre, University College Cork, Cork, Ireland
| | - Dominic G Roy
- Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Theresa Falls
- Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - John C Bell
- Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Mark Tangney
- Cork Cancer Research Centre, University College Cork, Cork, Ireland.
| |
Collapse
|
9
|
MDR-selective microbial-based therapy: a novel approach to cancer treatment. Med Hypotheses 2013; 81:207-11. [PMID: 23719029 DOI: 10.1016/j.mehy.2013.05.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 04/23/2013] [Accepted: 05/02/2013] [Indexed: 12/15/2022]
Abstract
Microbial-based therapy of cancer is one of the earliest non-surgical anticancer therapies. The main limitation of such therapies is the toxicity of the therapeutic dose. This article discusses a novel approach that exploits cancer multidrug resistance (MDR) to provide a safer microbial-based therapy. As multidrug resistant cells can only contain limited amounts of a variety of susceptible drugs including certain antibiotics, we can take advantage of MDR to create a micro-environment (antibiotic free) that favors growth of intracellular bacteria within cancer cells. Thus, this approach targets cancer cells and spares normal cells (shielded by antibiotic): providing a more selective thus safer anticancer treatment. This article also explores the potentials of Chlamydia pneumoniae as an anti-cancer agent in this MDR-selective microbial-based therapy: its unique life cycle and the immune response to its infection suggest that it could be used directly, in the proposed approach, without any pre-requirements.
Collapse
|
10
|
PEGylation of vesicular stomatitis virus extends virus persistence in blood circulation of passively immunized mice. J Virol 2013; 87:3752-9. [PMID: 23325695 DOI: 10.1128/jvi.02832-12] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We are developing oncolytic vesicular stomatitis viruses (VSVs) for systemic treatment of multiple myeloma, an incurable malignancy of antibody-secreting plasma cells that are specifically localized in the bone marrow. One of the presumed advantages for using VSV as an oncolytic virus is that human infections are rare and preexisting anti-VSV immunity is typically lacking in cancer patients, which is very important for clinical success. However, our studies show that nonimmune human and mouse serum can neutralize clinical-grade VSV, reducing the titer by up to 4 log units in 60 min. In addition, we show that neutralizing anti-VSV antibodies negate the antitumor efficacy of VSV, a concern for repeat VSV administration. We have investigated the potential use of covalent modification of VSV with polyethylene glycol (PEG) or a function-spacer-lipid (FSL)-PEG construct to inhibit serum neutralization and to limit hepatosplenic sequestration of systemically delivered VSV. We report that in mice passively immunized with neutralizing anti-VSV antibodies, PEGylation of VSV improved the persistence of VSV in the blood circulation, maintaining a more than 1-log-unit increase in VSV genome copies for up to 1 h compared to the genome copy numbers for the non-PEGylated virus, which was mostly cleared within 10 min after intravenous injection. We are currently investigating if this increase in PEGylated VSV circulating half-life can translate to increased virus delivery and better efficacy in mouse models of multiple myeloma.
Collapse
|
11
|
Buckingham L. A Look to the Future. Pharmacogenomics 2013. [DOI: 10.1016/b978-0-12-391918-2.00014-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|