1
|
Zinovieva M, Ryapolova A, Karabelsky A, Minskaia E. Oncolytic Vesicular Stomatitis Virus: Optimisation Strategies for Anti-Cancer Therapies. FRONT BIOSCI-LANDMRK 2024; 29:374. [PMID: 39614430 DOI: 10.31083/j.fbl2911374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 07/22/2024] [Accepted: 07/31/2024] [Indexed: 12/01/2024]
Abstract
Oncolytic viruses (OVs) represent a targeted anti-cancer therapy approach due to their ability not only to selectively infect and destroy malignant cells but also to induce an immune response. Vesicular stomatitis virus (VSV) offers a promising platform due to its low prevalence and pathogenicity in humans, lack of pre-existing immunity, easily manipulated genome, rapid growth to high titers in a broad range of cell lines, and inability to integrate into the host genome. However, despite its many advantages, many unresolved problems remain: problematic production based on the reverse genetics system, oncological selectivity, and the overall effectiveness of VSV monotherapy. This review will discuss various attempts at viral genome modifications aimed at improving the oncolytic properties of VSV. These strategies include inhibition of viral genes, modification of genes responsible for targeting cancer cells over healthy ones, insertion of foreign genes for boosting immune response, and changing the order of viral and inserted foreign genes. In addition, possible ways to improve VSV-based anti-tumor therapy and achieve higher efficiency will be considered by evaluating the effectiveness of various delivery methods as well as discussing treatment options by combining VSV with other groups of anticancer drugs.
Collapse
Affiliation(s)
- Margarita Zinovieva
- Department of Gene Therapy, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Anastasia Ryapolova
- Department of Gene Therapy, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Alexander Karabelsky
- Department of Gene Therapy, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Ekaterina Minskaia
- Department of Gene Therapy, Sirius University of Science and Technology, 354340 Sochi, Russia
| |
Collapse
|
2
|
Rangsitratkul C, Lawson C, Bernier-Godon F, Niavarani SR, Boudaud M, Rouleau S, Gladu-Corbin AO, Surendran A, Ekindi-Ndongo N, Koti M, Ilkow CS, Richard PO, Tai LH. Intravesical immunotherapy with a GM-CSF armed oncolytic vesicular stomatitis virus improves outcome in bladder cancer. Mol Ther Oncolytics 2022; 24:507-521. [PMID: 35229029 PMCID: PMC8851153 DOI: 10.1016/j.omto.2022.01.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 01/27/2022] [Indexed: 11/10/2022] Open
Abstract
A significant proportion of non-muscle invasive bladder cancer cases will progress to muscle invasive disease. Transurethral resection followed by Bacillus Calmette Guerin immunotherapy can reduce this risk, while cystectomy prior to muscle invasion provides the best option for survival. Currently, there are no effective treatments for Bacillus Calmette Guerin refractory disease. A novel oncolytic vesicular stomatitis virus containing the human GM-CSF transgene (VSVd51-hGM-CSF) was rescued and tested as a potential bladder-sparing therapy for aggressive bladder cancer. The existing variant expressing mouse GM-CSF was also used. Measurement of gene expression and protein level alterations of canonical immunogenic cell death associated events on mouse and human bladder cancer cell lines and spheroids showed enhanced release of danger signals and immunogenic factors following infection with VSVd51-m/hGM-CSF. Intravesical instillation of VSVd51-mGM-CSF into MB49 bladder cancer bearing C57Bl/6 mice demonstrated enhanced activation of peripheral and bladder infiltrating effector immune cells, along with improved survival and reduced tumor volume. Importantly, virus-mediated anti-tumor immunity was recapitulated in bladder cancer patient-derived organoids. These results suggest that VSVd51-hGM-CSF is a promising viro/immunotherapy that could benefit bladder cancer patients.
Collapse
|
3
|
Kim TE, Puckett S, Zhang K, Herpai DM, Ornelles DA, Davis JN, van den Pol AN, Debinski W, Lyles DS. Diversity in responses to oncolytic Lassa-vesicular stomatitis virus in patient-derived glioblastoma cells. MOLECULAR THERAPY-ONCOLYTICS 2021; 22:232-244. [PMID: 34514102 PMCID: PMC8424128 DOI: 10.1016/j.omto.2021.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 06/08/2021] [Indexed: 11/29/2022]
Abstract
The difficulty of glioblastoma treatment makes it a good candidate for novel therapies, such as oncolytic viruses. Vesicular stomatitis virus expressing Lassa virus glycoprotein (Lassa-VSV) showed significant promise in animal models using established glioblastoma cell lines. These experiments were to determine the susceptibility of low-passage, patient-derived cell lines to Lassa-VSV oncolysis. Four patient-derived glioblastoma cell lines were infected with Lassa-VSV that expresses green fluorescent protein (GFP) and analyzed by fluorescence microscopy, flow cytometry, and cell viability assays. Cells were also analyzed as tumorspheres containing primarily glioma stem-like cells. Three low-passage, patient-derived cells were further analyzed with RNA sequencing (RNA-seq). Individual cell lines varied somewhat in their levels of viral gene expression and time course of Lassa-VSV-induced cell death, but each was susceptible to Lassa-VSV. Brain Tumor Center of Excellence (BTCOE) 4765 cells had the highest level of expression of interferon-stimulated genes but were most susceptible to Lassa-VSV-induced cell death, indicating that more susceptible cells do not necessarily have lower interferon pathway activation. Cells cultured as tumorspheres and infected with Lassa-VSV also showed variable susceptibility to Lassa-VSV, but BTCOE 4765 cells were least susceptible. Thus, patient-derived brain tumor cells show variable responses to Lassa-VSV infection, but each of the lines was susceptible to VSV oncolysis.
Collapse
Affiliation(s)
- Teddy E Kim
- Department of Neurosurgery, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Shelby Puckett
- Department of Biochemistry, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157, USA
| | - Kailong Zhang
- Department of Biochemistry, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157, USA
| | - Denise M Herpai
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - David A Ornelles
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - John N Davis
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, USA
| | | | - Waldemar Debinski
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Douglas S Lyles
- Department of Biochemistry, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157, USA
| |
Collapse
|
4
|
Boesch M, Baty F, Rothschild SI, Tamm M, Joerger M, Früh M, Brutsche MH. Tumour neoantigen mimicry by microbial species in cancer immunotherapy. Br J Cancer 2021; 125:313-323. [PMID: 33824481 PMCID: PMC8329167 DOI: 10.1038/s41416-021-01365-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 02/02/2021] [Accepted: 03/10/2021] [Indexed: 02/08/2023] Open
Abstract
Tumour neoantigens arising from cancer-specific mutations generate a molecular fingerprint that has a definite specificity for cancer. Although this fingerprint perfectly discriminates cancer from healthy somatic and germline cells, and is therefore therapeutically exploitable using immune checkpoint blockade, gut and extra-gut microbial species can independently produce epitopes that resemble tumour neoantigens as part of their natural gene expression programmes. Such tumour molecular mimicry is likely not only to influence the quality and strength of the body's anti-cancer immune response, but could also explain why certain patients show favourable long-term responses to immune checkpoint blockade while others do not benefit at all from this treatment. This article outlines the requirement for tumour neoantigens in successful cancer immunotherapy and draws attention to the emerging role of microbiome-mediated tumour neoantigen mimicry in determining checkpoint immunotherapy outcome, with far-reaching implications for the future of cancer immunotherapy.
Collapse
Affiliation(s)
| | - Florent Baty
- Lung Center, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Sacha I Rothschild
- Department of Medical Oncology and Comprehensive Cancer Center, University Hospital of Basel, Basel, Switzerland
| | - Michael Tamm
- Department of Pulmonology, University Hospital of Basel, Basel, Switzerland
| | - Markus Joerger
- Department of Medical Oncology and Hematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Martin Früh
- Department of Medical Oncology and Hematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
- Department of Medical Oncology, University Hospital Bern, Bern, Switzerland
| | | |
Collapse
|
5
|
Kottke T, Tonne J, Evgin L, Driscoll CB, van Vloten J, Jennings VA, Huff AL, Zell B, Thompson JM, Wongthida P, Pulido J, Schuelke MR, Samson A, Selby P, Ilett E, McNiven M, Roberts LR, Borad MJ, Pandha H, Harrington K, Melcher A, Vile RG. Oncolytic virotherapy induced CSDE1 neo-antigenesis restricts VSV replication but can be targeted by immunotherapy. Nat Commun 2021; 12:1930. [PMID: 33772027 PMCID: PMC7997928 DOI: 10.1038/s41467-021-22115-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 02/25/2021] [Indexed: 01/06/2023] Open
Abstract
In our clinical trials of oncolytic vesicular stomatitis virus expressing interferon beta (VSV-IFNβ), several patients achieved initial responses followed by aggressive relapse. We show here that VSV-IFNβ-escape tumors predictably express a point-mutated CSDE1P5S form of the RNA-binding Cold Shock Domain-containing E1 protein, which promotes escape as an inhibitor of VSV replication by disrupting viral transcription. Given time, VSV-IFNβ evolves a compensatory mutation in the P/M Inter-Genic Region which rescues replication in CSDE1P5S cells. These data show that CSDE1 is a major cellular co-factor for VSV replication. However, CSDE1P5S also generates a neo-epitope recognized by non-tolerized T cells. We exploit this predictable neo-antigenesis to drive, and trap, tumors into an escape phenotype, which can be ambushed by vaccination against CSDE1P5S, preventing tumor escape. Combining frontline therapy with escape-targeting immunotherapy will be applicable across multiple therapies which drive tumor mutation/evolution and simultaneously generate novel, targetable immunopeptidomes associated with acquired treatment resistance.
Collapse
Affiliation(s)
- Timothy Kottke
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jason Tonne
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Laura Evgin
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Jacob van Vloten
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Victoria A Jennings
- Chester Beatty Laboratories, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
- Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Amanda L Huff
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Brady Zell
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jill M Thompson
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Jose Pulido
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Adel Samson
- Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Peter Selby
- Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Elizabeth Ilett
- Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Mark McNiven
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Lewis R Roberts
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Mitesh J Borad
- Division of Hematology/Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Hardev Pandha
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Kevin Harrington
- Chester Beatty Laboratories, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | - Alan Melcher
- Chester Beatty Laboratories, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | - Richard G Vile
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA.
- Leeds Institute of Medical Research, University of Leeds, Leeds, UK.
- Department of Immunology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
6
|
Lassa-VSV chimeric virus targets and destroys human and mouse ovarian cancer by direct oncolytic action and by initiating an anti-tumor response. Virology 2020; 555:44-55. [PMID: 33453650 DOI: 10.1016/j.virol.2020.10.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/18/2020] [Accepted: 10/28/2020] [Indexed: 11/24/2022]
Abstract
Ovarian cancer is the third most common female cancer, with poor survival in later stages of metastatic spread. We test a chimeric virus consisting of genes from Lassa and vesicular stomatitis viruses, LASV-VSV; the native VSV glycoprotein is replaced by the Lassa glycoprotein, greatly reducing neurotropism. Human ovarian cancer cells in immunocompromised nude mice were lethal in controls. Chemotherapeutic paclitaxel and cisplatin showed modest cancer inhibition and survival extension. In contrast, a single intraperitoneal injection of LASV-VSV selectively infected and killed ovarian cancer cells, generating long-term survival. Mice with human ovarian cancer cells in brain showed rapid deterioration; LASV-VSV microinjection into brain blocked cancer growth, and generated long-term survival. Treatment of immunocompetent mice with infected mouse ovarian cancer cells blocked growth of non-infected ovarian cancer cells peritoneally and in brain. These results suggest LASV-VSV is a viable candidate for further study and may be of use in the treatment of ovarian cancer.
Collapse
|
7
|
The Oncolytic Virus VSV-GP Is Effective against Malignant Melanoma. Viruses 2018; 10:v10030108. [PMID: 29498639 PMCID: PMC5869501 DOI: 10.3390/v10030108] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 02/12/2018] [Accepted: 02/24/2018] [Indexed: 12/12/2022] Open
Abstract
Previously, we described VSV-GP, a modified version of the vesicular stomatitis virus, as a non-neurotoxic oncolytic virus that is effective for the treatment of malignant glioblastoma and ovarian cancer. Here, we evaluate the therapeutic efficacy of VSV-GP for malignant melanoma. All of the human, mouse, and canine melanoma cell lines that were tested, alongside most primary human melanoma cultures, were infected by VSV-GP and efficiently killed. Additionally, we found that VSV-GP prolonged the survival of mice in both a xenograft and a syngeneic mouse model. However, only a few mice survived with long-term tumor remission. When we analyzed the factors that might limit VSV-GP's efficacy, we found that vector-neutralizing antibodies did not play a role in this context, as even after eight subsequent immunizations and an observation time of 42 weeks, no vector-neutralizing antibodies were induced in VSV-GP immunized mice. In contrast, the type I IFN response might have contributed to the reduced efficacy of the therapy, as both of the cell lines that were used for the mouse models were able to mount a protective IFN response. Nevertheless, early treatment with VSV-GP also reduced the number and size of lung metastases in a syngeneic B16 mouse model. In summary, VSV-GP is a potent candidate for the treatment of malignant melanoma; however, factors limiting the efficacy of the virus need to be further explored.
Collapse
|
8
|
Felt SA, Grdzelishvili VZ. Recent advances in vesicular stomatitis virus-based oncolytic virotherapy: a 5-year update. J Gen Virol 2017; 98:2895-2911. [PMID: 29143726 DOI: 10.1099/jgv.0.000980] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Oncolytic virus (OV) therapy is an anti-cancer approach that uses viruses that preferentially infect, replicate in and kill cancer cells. Vesicular stomatitis virus (VSV, a rhabdovirus) is an OV that is currently being tested in the USA in several phase I clinical trials against different malignancies. Several factors make VSV a promising OV: lack of pre-existing human immunity against VSV, a small and easy to manipulate genome, cytoplasmic replication without risk of host cell transformation, independence of cell cycle and rapid growth to high titres in a broad range of cell lines facilitating large-scale virus production. While significant advances have been made in VSV-based OV therapy, room for improvement remains. Here we review recent studies (published in the last 5 years) that address 'old' and 'new' challenges of VSV-based OV therapy. These studies focused on improving VSV safety, oncoselectivity and oncotoxicity; breaking resistance of some cancers to VSV; preventing premature clearance of VSV; and stimulating tumour-specific immunity. Many of these approaches were based on combining VSV with other therapeutics. This review also discusses another rhabdovirus closely related to VSV, Maraba virus, which is currently being tested in Canada in phase I/II clinical trials.
Collapse
Affiliation(s)
- Sébastien A Felt
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Valery Z Grdzelishvili
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA
| |
Collapse
|
9
|
Evaluation of the oncolytic potential of R 2B Mukteshwar vaccine strain of Newcastle disease virus (NDV) in a colon cancer cell line (SW-620). Arch Virol 2017; 162:2705-2713. [PMID: 28578522 DOI: 10.1007/s00705-017-3411-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 04/06/2017] [Indexed: 01/21/2023]
Abstract
Virotherapy is emerging as an alternative treatment of cancer. Among the candidate oncolytic viruses (OVs), Newcastle disease virus (NDV) has emerged as a promising non-engineered OV. In the present communication, we explored the oncolytic potential of R2B Mukteshwar strain of NDV using SW-620 colon cancer cells. SW-620 cells were xenografted in nude mice and after evaluation of the safety profile, 1 x 107 plaque forming units (PFU) of NDV were inoculated as virotherapeutic agent via the intratumoral (I/T) and intravenous (I/V) route. Tumor growth inhibition was compared with their respective control groups by gross volume and histopathological evaluation. Antibody titer and virus survival were measured by hemagglutination inhibition (HI)/serum neutralization test (SNT) and real-time PCR, respectively. During the safety trial, the test strain did not produce any abnormal symptoms nor weight loss in BALB/c mice. Significant tumor lytic activity was evident when viruses were injected via the I/T route. There was a 43 and 57% tumor growth inhibition on absolute and relative tumor volume basis, respectively, compared with mock control. On the same basis, the I/V route treatment resulted in 40 and 16% of inhibition, respectively. Histopathological examination revealed that the virus caused apoptosis, followed by necrosis, but immune cell infiltration was not remarkable. The virus survived in 2/2 mice until day 10 and in 3/6 mice by day 19, with both routes of administration. Anti-NDV antibodies were generated at moderate level and the titer reached a maximum of 1:32 and 1:64 via the I/T and I/V routes, respectively. In conclusion, the test NDV strain was found to be safe and showed oncolytic activity against the SW-620 cell line in mice.
Collapse
|
10
|
Shim KG, Zaidi S, Thompson J, Kottke T, Evgin L, Rajani KR, Schuelke M, Driscoll CB, Huff A, Pulido JS, Vile RG. Inhibitory Receptors Induced by VSV Viroimmunotherapy Are Not Necessarily Targets for Improving Treatment Efficacy. Mol Ther 2017; 25:962-975. [PMID: 28237836 DOI: 10.1016/j.ymthe.2017.01.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 01/26/2017] [Accepted: 01/26/2017] [Indexed: 12/28/2022] Open
Abstract
Systemic viroimmunotherapy activates endogenous innate and adaptive immune responses against both viral and tumor antigens. We have shown that therapy with vesicular stomatitis virus (VSV) engineered to express a tumor-associated antigen activates antigen-specific adoptively transferred T cells (adoptive cell therapy, ACT) in vivo to generate effective therapy. The overall goal of this study was to phenotypically characterize the immune response to VSV+ACT therapy and use the information gained to rationally improve combination therapy. We observed rapid expansion of blood CD8+ effector cells acutely following VSV therapy with markedly high expression of the immune checkpoint molecules PD-1 and TIM-3. Using these data, we tested a treatment schedule incorporating mAb immune checkpoint inhibitors with VSV+ACT treatment. Unlike clinical scenarios, we delivered therapy at early time points following tumor establishment and treatment. Our goal was to potentiate the immune response generated by VSV therapy to achieve durable control of metastatic disease. Despite the high frequency of endogenous PD-1+ TIM-3+ CD8+ T cells following virus administration, antibody blockade did not improve survival. These findings provide highly significant information about response kinetics to viroimmunotherapy and juxtapose the clinical use of checkpoint inhibitors against chronically dysfunctional T cells and the acute T cell response to oncolytic viruses.
Collapse
Affiliation(s)
- Kevin G Shim
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; Medical Scientist Training Program, Mayo Clinic, Rochester, MN 55905, USA
| | - Shane Zaidi
- Targeted Therapy Laboratory, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JJ, UK
| | - Jill Thompson
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Tim Kottke
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Laura Evgin
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Karishma R Rajani
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Matthew Schuelke
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; Medical Scientist Training Program, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Amanda Huff
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Jose S Pulido
- Department of Ophthalmology, Mayo Clinic, Rochester, MN 55905, USA
| | - Richard G Vile
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
11
|
Melzer MK, Lopez-Martinez A, Altomonte J. Oncolytic Vesicular Stomatitis Virus as a Viro-Immunotherapy: Defeating Cancer with a "Hammer" and "Anvil". Biomedicines 2017; 5:E8. [PMID: 28536351 PMCID: PMC5423493 DOI: 10.3390/biomedicines5010008] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 01/26/2017] [Accepted: 02/03/2017] [Indexed: 12/17/2022] Open
Abstract
Oncolytic viruses have gained much attention in recent years, due, not only to their ability to selectively replicate in and lyse tumor cells, but to their potential to stimulate antitumor immune responses directed against the tumor. Vesicular stomatitis virus (VSV), a negative-strand RNA virus, is under intense development as an oncolytic virus due to a variety of favorable properties, including its rapid replication kinetics, inherent tumor specificity, and its potential to elicit a broad range of immunomodulatory responses to break immune tolerance in the tumor microenvironment. Based on this powerful platform, a multitude of strategies have been applied to further improve the immune-stimulating potential of VSV and synergize these responses with the direct oncolytic effect. These strategies include: 1. modification of endogenous virus genes to stimulate interferon induction; 2. virus-mediated expression of cytokines or immune-stimulatory molecules to enhance anti-tumor immune responses; 3. vaccination approaches to stimulate adaptive immune responses against a tumor antigen; 4. combination with adoptive immune cell therapy for potentially synergistic therapeutic responses. A summary of these approaches will be presented in this review.
Collapse
Affiliation(s)
- Michael Karl Melzer
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany.
| | - Arturo Lopez-Martinez
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany.
| | - Jennifer Altomonte
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany.
| |
Collapse
|
12
|
Falls T, Roy DG, Bell JC, Bourgeois-Daigneault MC. Murine Tumor Models for Oncolytic Rhabdo-Virotherapy. ILAR J 2017; 57:73-85. [PMID: 27034397 DOI: 10.1093/ilar/ilv048] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The preclinical optimization and validation of novel treatments for cancer therapy requires the use of laboratory animals. Although in vitro experiments using tumor cell lines and ex vivo treatment of patient tumor samples provide a remarkable first-line tool for the initial study of tumoricidal potential, tumor-bearing animals remain the primary option to study delivery, efficacy, and safety of therapies in the context of a complete tumor microenvironment and functional immune system. In this review, we will describe the use of murine tumor models for oncolytic virotherapy using vesicular stomatitis virus. We will discuss studies using immunocompetent and immunodeficient models with respect to toxicity and therapeutic treatments, as well as the various techniques and tools available to study cancer therapy with Rhabdoviruses.
Collapse
Affiliation(s)
- Theresa Falls
- Theresa Falls is a research technician at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada. Dominic Guy Roy is a Ph.D candidate at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a Ph.D candidate in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. John Cameron Bell is a senior researcher at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and professor in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. Marie-Claude Bourgeois-Daigneault is a postdoctoral fellow at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a postdoctoral fellow in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada
| | - Dominic Guy Roy
- Theresa Falls is a research technician at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada. Dominic Guy Roy is a Ph.D candidate at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a Ph.D candidate in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. John Cameron Bell is a senior researcher at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and professor in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. Marie-Claude Bourgeois-Daigneault is a postdoctoral fellow at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a postdoctoral fellow in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada
| | - John Cameron Bell
- Theresa Falls is a research technician at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada. Dominic Guy Roy is a Ph.D candidate at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a Ph.D candidate in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. John Cameron Bell is a senior researcher at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and professor in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. Marie-Claude Bourgeois-Daigneault is a postdoctoral fellow at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a postdoctoral fellow in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada
| | - Marie-Claude Bourgeois-Daigneault
- Theresa Falls is a research technician at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada. Dominic Guy Roy is a Ph.D candidate at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a Ph.D candidate in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. John Cameron Bell is a senior researcher at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and professor in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. Marie-Claude Bourgeois-Daigneault is a postdoctoral fellow at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a postdoctoral fellow in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada
| |
Collapse
|
13
|
Abstract
Oncolytic viruses (OVs) comprise an emerging cancer therapeutic modality whose activity involves both direct tumor cell lysis and the induction of immunogenic cell death (ICD). Cellular proteins released from the OV-lysed tumor cells, known as damage-associated molecular patterns and tumor-associated antigens, activate dendritic cells and elicit adaptive antitumor immunity. Interaction with the innate immune system and the development of long-lasting immune memory also contribute to OV-induced cell death. The degree to which the ICD component contributes to the clinical efficacy of OV therapy is still unclear. Modulation of a range of immune interactions may be beneficial or detrimental in nature and the interactions depend on the specific tumor, the site and extent of the disease, the immunosuppressive tumor microenvironment, the OV platform, the dose, time, and delivery conditions, as well as individual patient responses. To enhance the contribution of ICD, OVs have been engineered to express immunostimulatory genes and strategies have been developed to combine OV therapy with chemo- and immune-based therapeutic regimens. However, these approaches carry the risk that they may also be tolerogenic depending on their levels and the presence of other cytokines, their direct antiviral effects, and the timing and conditions of their expression. The contribution of autophagy to adaptive immunity, the ability of the OVs to kill cancer stem cells, and the patient’s baseline immune status are additional considerations. This review focuses on the complex and as yet poorly understood balancing act that dictates the outcome of OV therapy. We summarize current understanding of the OVs’ function in eliciting antitumor immunity and its relationship to therapeutic efficacy. Also discussed are the criteria involved in restraining antiviral immune responses and minimizing pathology while promoting antitumor immunity to override immune tolerance.
Collapse
Affiliation(s)
- Laure Aurelian
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
14
|
Allan KJ, Stojdl DF, Swift SL. High-throughput screening to enhance oncolytic virus immunotherapy. Oncolytic Virother 2016; 5:15-25. [PMID: 27579293 PMCID: PMC4996253 DOI: 10.2147/ov.s66217] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
High-throughput screens can rapidly scan and capture large amounts of information across multiple biological parameters. Although many screens have been designed to uncover potential new therapeutic targets capable of crippling viruses that cause disease, there have been relatively few directed at improving the efficacy of viruses that are used to treat disease. Oncolytic viruses (OVs) are biotherapeutic agents with an inherent specificity for treating malignant disease. Certain OV platforms – including those based on herpes simplex virus, reovirus, and vaccinia virus – have shown success against solid tumors in advanced clinical trials. Yet, many of these OVs have only undergone minimal engineering to solidify tumor specificity, with few extra modifications to manipulate additional factors. Several aspects of the interaction between an OV and a tumor-bearing host have clear value as targets to improve therapeutic outcomes. At the virus level, these include delivery to the tumor, infectivity, productivity, oncolysis, bystander killing, spread, and persistence. At the host level, these include engaging the immune system and manipulating the tumor microenvironment. Here, we review the chemical- and genome-based high-throughput screens that have been performed to manipulate such parameters during OV infection and analyze their impact on therapeutic efficacy. We further explore emerging themes that represent key areas of focus for future research.
Collapse
Affiliation(s)
- K J Allan
- Children's Hospital of Eastern Ontario (CHEO) Research Institute; Department of Biology, Microbiology and Immunology
| | - David F Stojdl
- Children's Hospital of Eastern Ontario (CHEO) Research Institute; Department of Biology, Microbiology and Immunology; Department of Pediatrics, University of Ottawa, Ottawa, ON, Canada
| | - S L Swift
- Children's Hospital of Eastern Ontario (CHEO) Research Institute
| |
Collapse
|
15
|
Janelle V, Lamarre A. How Informative is the Immune Response Against Surrogate Tumor Antigens to Assess Antitumor Immunity? Front Oncol 2014; 4:135. [PMID: 24926437 PMCID: PMC4045314 DOI: 10.3389/fonc.2014.00135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 05/21/2014] [Indexed: 11/13/2022] Open
Affiliation(s)
- Valérie Janelle
- Immunovirology Laboratory, Institut National de la Recherche Scientifique, INRS-Institut Armand-Frappier , Laval, QC , Canada ; Department of Biology, Biomed Research Center, Université du Québec à Montréal , Montréal, QC , Canada
| | - Alain Lamarre
- Immunovirology Laboratory, Institut National de la Recherche Scientifique, INRS-Institut Armand-Frappier , Laval, QC , Canada ; Department of Biology, Biomed Research Center, Université du Québec à Montréal , Montréal, QC , Canada
| |
Collapse
|
16
|
Guo ZS, Liu Z, Bartlett DL. Oncolytic Immunotherapy: Dying the Right Way is a Key to Eliciting Potent Antitumor Immunity. Front Oncol 2014; 4:74. [PMID: 24782985 PMCID: PMC3989763 DOI: 10.3389/fonc.2014.00074] [Citation(s) in RCA: 214] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 03/24/2014] [Indexed: 12/22/2022] Open
Abstract
Oncolytic viruses (OVs) are novel immunotherapeutic agents whose anticancer effects come from both oncolysis and elicited antitumor immunity. OVs induce mostly immunogenic cancer cell death (ICD), including immunogenic apoptosis, necrosis/necroptosis, pyroptosis, and autophagic cell death, leading to exposure of calreticulin and heat-shock proteins to the cell surface, and/or released ATP, high-mobility group box 1, uric acid, and other damage-associated molecular patterns as well as pathogen-associated molecular patterns as danger signals, along with tumor-associated antigens, to activate dendritic cells and elicit adaptive antitumor immunity. Dying the right way may greatly potentiate adaptive antitumor immunity. The mode of cancer cell death may be modulated by individual OVs and cancer cells as they often encode and express genes that inhibit/promote apoptosis, necroptosis, or autophagic cell death. We can genetically engineer OVs with death-pathway-modulating genes and thus skew the infected cancer cells toward certain death pathways for the enhanced immunogenicity. Strategies combining with some standard therapeutic regimens may also change the immunological consequence of cancer cell death. In this review, we discuss recent advances in our understanding of danger signals, modes of cancer cell death induced by OVs, the induced danger signals and functions in eliciting subsequent antitumor immunity. We also discuss potential combination strategies to target cells into specific modes of ICD and enhance cancer immunogenicity, including blockade of immune checkpoints, in order to break immune tolerance, improve antitumor immunity, and thus the overall therapeutic efficacy.
Collapse
Affiliation(s)
- Zong Sheng Guo
- Department of Surgery, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine , Pittsburgh, PA , USA
| | - Zuqiang Liu
- Department of Surgery, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine , Pittsburgh, PA , USA
| | - David L Bartlett
- Department of Surgery, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine , Pittsburgh, PA , USA
| |
Collapse
|