1
|
Jackson Cullison SR, Flemming JP, Karagoz K, Wermuth PJ, Mahoney MG. Mechanisms of extracellular vesicle uptake and implications for the design of cancer therapeutics. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e70017. [PMID: 39483807 PMCID: PMC11522837 DOI: 10.1002/jex2.70017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/11/2024] [Accepted: 10/14/2024] [Indexed: 11/03/2024]
Abstract
The translation of pre-clinical anti-cancer therapies to regulatory approval has been promising, but slower than hoped. While innovative and effective treatments continue to achieve or seek approval, setbacks are often attributed to a lack of efficacy, failure to achieve clinical endpoints, and dose-limiting toxicities. Successful efforts have been characterized by the development of therapeutics designed to specifically deliver optimal and effective dosing to tumour cells while minimizing off-target toxicity. Much effort has been devoted to the rational design and application of synthetic nanoparticles to serve as targeted therapeutic delivery vehicles. Several challenges to the successful application of this modality as delivery vehicles include the induction of a protracted immune response that results in their rapid systemic clearance, manufacturing cost, lack of stability, and their biocompatibility. Extracellular vesicles (EVs) are a heterogeneous class of endogenous biologically produced lipid bilayer nanoparticles that mediate intercellular communication by carrying bioactive macromolecules capable of modifying cellular phenotypes to local and distant cells. By genetic, chemical, or metabolic methods, extracellular vesicles (EVs) can be engineered to display targeting moieties on their surface while transporting specific cargo to modulate pathological processes following uptake by target cell populations. This review will survey the types of EVs, their composition and cargoes, strategies employed to increase their targeting, uptake, and cargo release, and their potential as targeted anti-cancer therapeutic delivery vehicles.
Collapse
Affiliation(s)
| | - Joseph P. Flemming
- Rowan‐Virtua School of Osteopathic MedicineRowan UniversityStratfordNew JerseyUSA
| | - Kubra Karagoz
- Departments of PharmacologyPhysiology, and Cancer Biology, Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | | | - Mỹ G. Mahoney
- Departments of PharmacologyPhysiology, and Cancer Biology, Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
- Department of Otolaryngology – Head and Neck SurgeryThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
2
|
Metko M, Tonne J, Veliz Rios A, Thompson J, Mudrick H, Masopust D, Diaz RM, Barry MA, Vile RG. Intranasal Prime-Boost with Spike Vectors Generates Antibody and T-Cell Responses at the Site of SARS-CoV-2 Infection. Vaccines (Basel) 2024; 12:1191. [PMID: 39460356 PMCID: PMC11511174 DOI: 10.3390/vaccines12101191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Long-lived, re-activatable immunity to SARS-CoV-2 and its emerging variants will rely on T cells recognizing conserved regions of viral proteins across strains. Heterologous prime-boost regimens can elicit elevated levels of circulating CD8+ T cells that provide a reservoir of first responders upon viral infection. Although most vaccines are currently delivered intramuscularly (IM), the initial site of infection is the nasal cavity. METHODS Here, we tested the hypothesis that a heterologous prime and boost vaccine regimen delivered intranasally (IN) will generate improved immune responses locally at the site of virus infection compared to intramuscular vaccine/booster regimens. RESULTS In a transgenic human ACE2 murine model, both a Spike-expressing single-cycle adenovirus (SC-Ad) and an IFNß safety-enhanced replication-competent Vesicular Stomatitis Virus (VSV) platform generated anti-Spike antibody and T-cell responses that diminished with age. Although SC-Ad-Spike boosted a prime with VSV-Spike-mIFNß, SC-Ad-Spike alone induced maximal levels of IgG, IgA, and CD8+ T-cell responses. CONCLUSIONS There were significant differences in T-cell responses in spleens compared to lungs, and the intranasal boost was significantly superior to the intramuscular boost in generating sentinel immune effectors at the site of the virus encounter in the lungs. These data show that serious consideration should be given to intranasal boosting with anti-SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Muriel Metko
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (M.M.); (J.T.); (A.V.R.); (J.T.); (R.M.D.)
| | - Jason Tonne
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (M.M.); (J.T.); (A.V.R.); (J.T.); (R.M.D.)
| | - Alexa Veliz Rios
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (M.M.); (J.T.); (A.V.R.); (J.T.); (R.M.D.)
| | - Jill Thompson
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (M.M.); (J.T.); (A.V.R.); (J.T.); (R.M.D.)
| | - Haley Mudrick
- Molecular Pharmacology and Experimental Therapeutics Program, Mayo Clinic, Rochester, MN 55905, USA;
| | - David Masopust
- Department of Microbiology & Immunology, University of Minnesota Medical School, 2101 6th St. SE, Minneapolis, MN 55455, USA;
| | - Rosa Maria Diaz
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (M.M.); (J.T.); (A.V.R.); (J.T.); (R.M.D.)
| | - Michael A. Barry
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (M.M.); (J.T.); (A.V.R.); (J.T.); (R.M.D.)
- Department of Infectious Diseases, Mayo Clinic, Rochester, MN 55905, USA
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Richard G. Vile
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (M.M.); (J.T.); (A.V.R.); (J.T.); (R.M.D.)
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
3
|
Webb MJ, Sangsuwannukul T, van Vloten J, Evgin L, Kendall B, Tonne J, Thompson J, Metko M, Moore M, Chiriboga Yerovi MP, Olin M, Borgatti A, McNiven M, Monga SPS, Borad MJ, Melcher A, Roberts LR, Vile R. Expression of tumor antigens within an oncolytic virus enhances the anti-tumor T cell response. Nat Commun 2024; 15:5442. [PMID: 38937436 PMCID: PMC11211353 DOI: 10.1038/s41467-024-49286-x] [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: 11/07/2023] [Accepted: 05/29/2024] [Indexed: 06/29/2024] Open
Abstract
Although patients benefit from immune checkpoint inhibition (ICI) therapy in a broad variety of tumors, resistance may arise from immune suppressive tumor microenvironments (TME), which is particularly true of hepatocellular carcinoma (HCC). Since oncolytic viruses (OV) can generate a highly immune-infiltrated, inflammatory TME, OVs could potentially restore ICI responsiveness via recruitment, priming, and activation of anti-tumor T cells. Here we find that on the contrary, an oncolytic vesicular stomatitis virus, expressing interferon-ß (VSV-IFNß), antagonizes the effect of anti-PD-L1 therapy in a partially anti-PD-L1-responsive model of HCC. Cytometry by Time of Flight shows that VSV-IFNß expands dominant anti-viral effector CD8 T cells with concomitant relative disappearance of anti-tumor T cell populations, which are the target of anti-PD-L1. However, by expressing a range of HCC tumor antigens within VSV, combination OV and anti-PD-L1 therapeutic benefit could be restored. Our data provide a cautionary message for the use of highly immunogenic viruses as tumor-specific immune-therapeutics by showing that dominant anti-viral T cell responses can inhibit sub-dominant anti-tumor T cell responses. However, through encoding tumor antigens within the virus, oncolytic virotherapy can generate anti-tumor T cell populations upon which immune checkpoint blockade can effectively work.
Collapse
Affiliation(s)
- Mason J Webb
- Department of Hematology/Medical Oncology, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | | | - Jacob van Vloten
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Laura Evgin
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, V5Z1L3, Canada
- Michael Smith Genome Sciences Department, BC Cancer Research Institute, Vancouver, BC, V5Z1L3, Canada
| | - Benjamin Kendall
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jason Tonne
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jill Thompson
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Muriel Metko
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Madelyn Moore
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA
| | | | - Michael Olin
- Division of Pediatric Hematology and Oncology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Antonella Borgatti
- Department of Veterinary Clinical Sciences, University of Minnesota, St. Paul, MN, 55108, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA
- Clinical Investigation Center, University of Minnesota, St. Paul, MN, 55108, USA
| | - Mark McNiven
- Mayo Center for Biomedical Discovery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Satdarshan P S Monga
- Pittsburgh Liver Institute, University of Pittsburgh and UPMC, Pittsburgh, PA, 15261, USA
| | - Mitesh J Borad
- Department of Hematology/Medical Oncology, Mayo Clinic, Phoenix, AZ, 85054, USA
| | - Alan Melcher
- Division of Radiotherapy and Imaging, Institute of Cancer Research, Chester Beatty Laboratories, London, SW3 6JB, UK
| | - Lewis R Roberts
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Richard Vile
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Immunology, Mayo Clinic, Rochester, MN, 55905, USA.
- Joan Reece Department of Immuno-oncology, King's College London, London, UK.
| |
Collapse
|
4
|
Makielski KM, Sarver AL, Henson MS, Stuebner KM, Borgatti A, Suksanpaisan L, Preusser C, Tabaran AF, Cornax I, O’Sullivan MG, Chehadeh A, Groschen D, Bergsrud K, Pracht S, Winter A, Mills LJ, Schwabenlander MD, Wolfe M, Farrar MA, Cutter GR, Koopmeiners JS, Russell SJ, Modiano JF, Naik S. Neoadjuvant systemic oncolytic vesicular stomatitis virus is safe and may enhance long-term survivorship in dogs with naturally occurring osteosarcoma. Mol Ther Oncolytics 2023; 31:100736. [PMID: 37965295 PMCID: PMC10641240 DOI: 10.1016/j.omto.2023.100736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 10/11/2023] [Indexed: 11/16/2023] Open
Abstract
Osteosarcoma is a devastating bone cancer that disproportionally afflicts children, adolescents, and young adults. Standard therapy includes surgical tumor resection combined with multiagent chemotherapy, but many patients still suffer from metastatic disease progression. Neoadjuvant systemic oncolytic virus (OV) therapy has the potential to improve clinical outcomes by targeting primary and metastatic tumor sites and inducing durable antitumor immune responses. Here we describe the first evaluation of neoadjuvant systemic therapy with a clinical-stage recombinant oncolytic vesicular stomatitis virus (VSV), VSV-IFNβ-NIS, in naturally occurring cancer, specifically appendicular osteosarcoma in companion dogs. Canine osteosarcoma has a similar natural disease history as its human counterpart. VSV-IFNβ-NIS was administered prior to standard of care surgical resection, permitting microscopic and genomic analysis of tumors. Treatment was well-tolerated and a "tail" of long-term survivors (∼35%) was apparent in the VSV-treated group, a greater proportion than observed in two contemporary control cohorts. An increase in tumor inflammation was observed in VSV-treated tumors and RNA-seq analysis showed that all the long-term responders had increased expression of a T cell anchored immune gene cluster. We conclude that neoadjuvant VSV-IFNβ-NIS is safe and may increase long-term survivorship in dogs with naturally occurring osteosarcoma, particularly those that exhibit pre-existing antitumor immunity.
Collapse
Affiliation(s)
- Kelly M. Makielski
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN 55108, USA
| | - Aaron L. Sarver
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Michael S. Henson
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN 55108, USA
| | - Kathleen M. Stuebner
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN 55108, USA
| | - Antonella Borgatti
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN 55108, USA
| | | | - Caitlin Preusser
- Department of Molecular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | | | - Ingrid Cornax
- Department of Veterinary Population Medicine, St. Paul, MN 55108, USA
| | | | - Andrea Chehadeh
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN 55108, USA
| | - Donna Groschen
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN 55108, USA
| | - Kelly Bergsrud
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN 55108, USA
| | - Sara Pracht
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN 55108, USA
| | - Amber Winter
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN 55108, USA
| | - Lauren J. Mills
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Pediatrics, University of Minnesota School of Medicine, Minneapolis, MN 55455, USA
| | - Marc D. Schwabenlander
- Veterinary Diagnostic Laboratory, University of Minnesota College of Veterinary Medicine, St. Paul, MN 55108, USA
| | - Melissa Wolfe
- Veterinary Diagnostic Laboratory, University of Minnesota College of Veterinary Medicine, St. Paul, MN 55108, USA
| | - Michael A. Farrar
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Gary R. Cutter
- University of Alabama at Birmingham, Birmingham, AL, USA
| | - Joseph S. Koopmeiners
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Division of Biostatistics, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Stephen J. Russell
- Department of Molecular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
- Vyriad, Inc., 2900 37th St NW, Rochester, MN 55901, USA
| | - Jaime F. Modiano
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN 55108, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Engineering and Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Shruthi Naik
- Department of Molecular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
- Vyriad, Inc., 2900 37th St NW, Rochester, MN 55901, USA
| |
Collapse
|
5
|
Vile R, Webb M, van Vloten J, Evgin L, Sangsuwannukul T, Kendall B, Tonne J, Thompson J, Metko M, Moore M, Yerovi MC, McNiven M, Monga S, Borad M, Roberts L. Chimerization of the Anti-Viral CD8 + T Cell Response with A Broad Anti-Tumor T Cell Response Reverses Inhibition of Checkpoint Blockade Therapy by Oncolytic Virotherapy. RESEARCH SQUARE 2023:rs.3.rs-3576281. [PMID: 38045348 PMCID: PMC10690324 DOI: 10.21203/rs.3.rs-3576281/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Although immune checkpoint inhibition (ICI) has produced profound survival benefits in a broad variety of tumors, a proportion of patients do not respond. Treatment failure is in part due to immune suppressive tumor microenvironments (TME), which is particularly true of hepatocellular carcinoma (HCC). Since oncolytic viruses (OV) can generate a highly immune-infiltrated, inflammatory TME, we developed a vesicular stomatitis virus expressing interferon-ß (VSV-IFNß) as a viro-immunotherapy against HCC. Since HCC standard of care atezolizumab/bevacizumab incorporates ICI, we tested the hypothesis that pro-inflammatory VSV-IFNß would recruit, prime, and activate anti-tumor T cells, whose activity anti-PD-L1 ICI would potentiate. However, in a partially anti-PD-L1-responsive model of HCC, addition of VSV-IFNß abolished anti-PD-L1 therapy. Cytometry by Time of Flight showed that VSV-IFNß expanded dominant anti-viral effector CD8 T cells with concomitant, relative disappearance of anti-tumor T cell populations which are the target of anti-PD-L1. However, by expressing a range of HCC tumor antigens within VSV, the potent anti-viral response became amalgamated with an anti-tumor T cell response generating highly significant cures compared to anti-PD-L1 ICI alone. Our data provide a cautionary message for the use of highly immunogenic viruses as tumor-specific immune-therapeutics by showing that dominant anti-viral T cell responses can inhibit sub-dominant anti-tumor T cell responses. However, by chimerizing anti-viral and anti-tumor T cell responses through encoding tumor antigens within the virus, oncolytic virotherapy can be purposed for very effective immune driven tumor clearance and can generate anti-tumor T cell populations upon which immune checkpoint blockade can effectively work.
Collapse
|
6
|
Smith KER, Peng KW, Pulido JS, Weisbrod AJ, Strand CA, Allred JB, Newsom AN, Zhang L, Packiriswamy N, Kottke T, Tonne JM, Moore M, Montane HN, Kottschade LA, McWilliams RR, Dudek AZ, Yan Y, Dimou A, Markovic SN, Federspiel MJ, Vile RG, Dronca RS, Block MS. A phase I oncolytic virus trial with vesicular stomatitis virus expressing human interferon beta and tyrosinase related protein 1 administered intratumorally and intravenously in uveal melanoma: safety, efficacy, and T cell responses. Front Immunol 2023; 14:1279387. [PMID: 38022659 PMCID: PMC10644866 DOI: 10.3389/fimmu.2023.1279387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Metastatic uveal melanoma (MUM) has a poor prognosis and treatment options are limited. These patients do not typically experience durable responses to immune checkpoint inhibitors (ICIs). Oncolytic viruses (OV) represent a novel approach to immunotherapy for patients with MUM. Methods We developed an OV with a Vesicular Stomatitis Virus (VSV) vector modified to express interferon-beta (IFN-β) and Tyrosinase Related Protein 1 (TYRP1) (VSV-IFNβ-TYRP1), and conducted a Phase 1 clinical trial with a 3 + 3 design in patients with MUM. VSV-IFNβ-TYRP1 was injected into a liver metastasis, then administered on the same day as a single intravenous (IV) infusion. The primary objective was safety. Efficacy was a secondary objective. Results 12 patients with previously treated MUM were enrolled. Median follow up was 19.1 months. 4 dose levels (DLs) were evaluated. One patient at DL4 experienced dose limiting toxicities (DLTs), including decreased platelet count (grade 3), increased aspartate aminotransferase (AST), and cytokine release syndrome (CRS). 4 patients had stable disease (SD) and 8 patients had progressive disease (PD). Interferon gamma (IFNγ) ELIspot data showed that more patients developed a T cell response to virus encoded TYRP1 at higher DLs, and a subset of patients also had a response to other melanoma antigens, including gp100, suggesting epitope spreading. 3 of the patients who responded to additional melanoma antigens were next treated with ICIs, and 2 of these patients experienced durable responses. Discussion Our study found that VSV-IFNβ -TYRP1 can be safely administered via intratumoral (IT) and IV routes in a previously treated population of patients with MUM. Although there were no clear objective radiographic responses to VSV-IFNβ-TYRP1, dose-dependent immunogenicity to TYRP1 and other melanoma antigens was seen.
Collapse
Affiliation(s)
| | - Kah-Whye Peng
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Jose S. Pulido
- Department of Ophthalmology, Wills Eye Hospital, Philadelphia, PA, United States
| | - Adam J. Weisbrod
- Department of Radiology, Mayo Clinic, Rochester, MN, United States
| | - Carrie A. Strand
- Department of Biostatistics and Informatics, Mayo Clinic, Rochester, MN, United States
| | - Jacob B. Allred
- Department of Biostatistics and Informatics, Mayo Clinic, Rochester, MN, United States
| | - Alysha N. Newsom
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Lianwen Zhang
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, United States
| | | | - Timothy Kottke
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Jason M. Tonne
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Madelyn Moore
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Heather N. Montane
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, United States
| | - Lisa A. Kottschade
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, United States
| | | | - Arkadiusz Z. Dudek
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, United States
| | - Yiyi Yan
- Department of Hematology and Oncology, Mayo Clinic Florida, Jacksonville, FL, United States
| | - Anastasios Dimou
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, United States
| | | | - Mark J. Federspiel
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Richard G. Vile
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Roxana S. Dronca
- Department of Hematology and Oncology, Mayo Clinic Florida, Jacksonville, FL, United States
| | - Matthew S. Block
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
7
|
Tur-Planells V, García-Sastre A, Cuadrado-Castano S, Nistal-Villan E. Engineering Non-Human RNA Viruses for Cancer Therapy. Vaccines (Basel) 2023; 11:1617. [PMID: 37897020 PMCID: PMC10611381 DOI: 10.3390/vaccines11101617] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Alongside the development and progress in cancer immunotherapy, research in oncolytic viruses (OVs) continues advancing novel treatment strategies to the clinic. With almost 50 clinical trials carried out over the last decade, the opportunities for intervention using OVs are expanding beyond the old-fashioned concept of "lytic killers", with promising breakthrough therapeutic strategies focused on leveraging the immunostimulatory potential of different viral platforms. This review presents an overview of non-human-adapted RNA viruses engineered for cancer therapy. Moreover, we describe the diverse strategies employed to manipulate the genomes of these viruses to optimize their therapeutic capabilities. By focusing on different aspects of this particular group of viruses, we describe the insights into the promising advancements in the field of virotherapy and its potential to revolutionize cancer treatment.
Collapse
Affiliation(s)
- Vicent Tur-Planells
- Microbiology Section, Department of Pharmaceutical Science and Health, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain;
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sara Cuadrado-Castano
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Genomics Institute (IGI), Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Estanislao Nistal-Villan
- Microbiology Section, Department of Pharmaceutical Science and Health, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain;
- Departamento de Ciencias Médicas Básicas, Instituto de Medicina Molecular Aplicada (IMMA) Nemesio Díez, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, 28668 Boadilla del Monte, Spain
| |
Collapse
|
8
|
Gao Y, Bergman I. Vesicular Stomatitis Virus (VSV) G Glycoprotein Can Be Modified to Create a Her2/Neu-Targeted VSV That Eliminates Large Implanted Mammary Tumors. J Virol 2023; 97:e0037223. [PMID: 37199666 PMCID: PMC10308914 DOI: 10.1128/jvi.00372-23] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 04/21/2023] [Indexed: 05/19/2023] Open
Abstract
Viral oncolytic immunotherapy is a nascent field that is developing tools to direct the immune system to find and eliminate cancer cells. Safety is improved by using cancer-targeted viruses that infect or grow poorly on normal cells. The recent discovery of the low-density lipoprotein (LDL) receptor as the major vesicular stomatitis virus (VSV) binding site allowed for the creation of a Her2/neu-targeted replicating recombinant VSV (rrVSV-G) by eliminating the LDL receptor binding site in the VSV-G glycoprotein (gp) and adding a sequence coding for a single chain antibody (SCA) to the Her2/neu receptor. The virus was adapted by serial passage on Her2/neu-expressing cancer cells resulting in a virus that yielded a 15- to 25-fold higher titer following in vitro infection of Her2/neu+-expressing cell lines than that of Her2/neu-negative cells (~1 × 108/mL versus 4 × 106 to 8 × 106/mL). An essential mutation resulting in a higher titer virus was a threonine-to-arginine change that produced an N-glycosylation site in the SCA. Infection of Her2/neu+ subcutaneous tumors yielded >10-fold more virus on days 1 and 2 than Her2/neu- tumors, and virus production continued for 5 days in Her2/neu+ tumors compared with 3 days that of 3 days in Her2/neu- tumors. rrVSV-G cured 70% of large 5-day peritoneal tumors compared with a 10% cure by a previously targeted rrVSV with a modified Sindbis gp. rrVSV-G also cured 33% of very large 7-day tumors. rrVSV-G is a new targeted oncolytic virus that has potent antitumor capabilities and allows for heterologous combination with other targeted oncolytic viruses. IMPORTANCE A new form of vesicular stomatitis virus (VSV) was created that specifically targets and destroys cancer cells that express the Her2/neu receptor. This receptor is commonly found in human breast cancer and is associated with a poor prognosis. In laboratory tests using mouse models, the virus was highly effective at eliminating implanted tumors and creating a strong immune response against cancer. VSV has many advantages as a cancer treatment, including high levels of safety and efficacy and the ability to be combined with other oncolytic viruses to enhance treatment results or to create an effective cancer vaccine. This new virus can also be easily modified to target other cancer cell surface molecules and to add immune-modifying genes. Overall, this new VSV is a promising candidate for further development as an immune-based cancer therapy.
Collapse
Affiliation(s)
- Yanhua Gao
- Department of Pediatrics, University of Pittsburgh School of Medicine, Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ira Bergman
- Department of Pediatrics, University of Pittsburgh School of Medicine, Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
9
|
Webb MJ, Kottke T, Kendall BL, Swanson J, Uzendu C, Tonne J, Thompson J, Metko M, Moore M, Borad M, Roberts L, Diaz RM, Olin M, Borgatti A, Vile R. Trap and ambush therapy using sequential primary and tumor escape-selective oncolytic viruses. Mol Ther Oncolytics 2023; 29:129-142. [PMID: 37313455 PMCID: PMC10258242 DOI: 10.1016/j.omto.2023.05.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 05/18/2023] [Indexed: 06/15/2023] Open
Abstract
In multiple models of oncolytic virotherapy, it is common to see an early anti-tumor response followed by recurrence. We have previously shown that frontline treatment with oncolytic VSV-IFN-β induces APOBEC proteins, promoting the selection of specific mutations that allow tumor escape. Of these mutations in B16 melanoma escape (ESC) cells, a C-T point mutation in the cold shock domain-containing E1 (CSDE1) gene was present at the highest frequency, which could be used to ambush ESC cells by vaccination with the mutant CSDE1 expressed within the virus. Here, we show that the evolution of viral ESC tumor cells harboring the escape-promoting CSDE1C-T mutation can also be exploited by a virological ambush. By sequential delivery of two oncolytic VSVs in vivo, tumors which would otherwise escape VSV-IFN-β oncolytic virotherapy could be cured. This also facilitated the priming of anti-tumor T cell responses, which could be further exploited using immune checkpoint blockade with the CD200 activation receptor ligand (CD200AR-L) peptide. Our findings here are significant in that they offer the possibility to develop oncolytic viruses as highly specific, escape-targeting viro-immunotherapeutic agents to be used in conjunction with recurrence of tumors following multiple different types of frontline cancer therapies.
Collapse
Affiliation(s)
- Mason J. Webb
- Division of Hematology/Medical Oncology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Timothy Kottke
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Jack Swanson
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Chisom Uzendu
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Jason Tonne
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Jill Thompson
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Muriel Metko
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Madelyn Moore
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Mitesh Borad
- Division of Hematology/Oncology, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Lewis Roberts
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Rosa M. Diaz
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Michael Olin
- Division of Pediatric Hematology and Oncology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Antonella Borgatti
- Department of Veterinary Clinical Sciences, University of Minnesota, St. Paul, MN 55108, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Clinical Investigation Center, University of Minnesota, St. Paul, MN 55108, USA
| | - Richard Vile
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
10
|
Raghuvanshi V, Yadav P, Ali S. Interferon production by Viral, Bacterial & Yeast system: A comparative overview in 2023. Int Immunopharmacol 2023; 120:110340. [PMID: 37230033 DOI: 10.1016/j.intimp.2023.110340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/19/2023] [Accepted: 05/11/2023] [Indexed: 05/27/2023]
Abstract
Interferons play a critical role in the innate immune response against several infections and play a key role in the control of a variety of viral and bacterial infectious diseases such as hepatitis, covid-19, cancer, and multiple sclerosis. Therefore, natural or synthetic IFN production is important and had three common methods, including bacterial fermentation, animal cell culture, and recombinant nucleic acid technology. However, the safety, purity, and accuracy of the most preferred INF production systems have not been extensively studied. This study provides a comprehensive comparative overview of interferon production in various systems that include viral, bacterial, yeast, and mammalian. We aim to determine the most efficient, safe, and accurate interferon production system available in the year 2023. The mechanisms of artificial interferon production were reviewed in various organisms, and the types and subtypes of interferons produced by each system were compared. Our analysis provides a comprehensive overview of the similarities and differences in interferon production and highlights the potential for developing new therapeutic strategies to combat infectious diseases. This review article offers the diverse strategies used by different organisms in producing and utilizing interferons, providing a framework for future research into the evolution and function of this critical immune response pathway.
Collapse
Affiliation(s)
| | - Pramod Yadav
- Research Assistant, Department of AFAF, Amity University Noida, Uttar Pradesh, 201313, India.
| | - Samim Ali
- Research Assistant, Kalpana Chawla Government Medical College Karnal, Haryana, 13200, India.
| |
Collapse
|
11
|
Moglan AM, Albaradie OA, Alsayegh FF, Alharbi HM, Samman YM, Jalal MM, Saeedi NH, Mahmoud AB, Alkayyal AA. Preclinical efficacy of oncolytic VSV-IFNβ in treating cancer: A systematic review. Front Immunol 2023; 14:1085940. [PMID: 37063914 PMCID: PMC10104167 DOI: 10.3389/fimmu.2023.1085940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 03/17/2023] [Indexed: 04/03/2023] Open
Abstract
BackgroundCancer incidence and mortality are increasing rapidly worldwide, necessitating further investigation into developing and optimizing emergent cancer therapies. Oncolytic viruses such as vesicular stomatitis virus encoding interferon β (VSV-IFNβ) have attracted considerable attention, as they offer great efficacy and safety profiles. This systematic review aimed to determine and compare the efficacy profile between VSV-IFNβ and non-treatment controls in preclinical cancer models.MethodologyThe Embase and Medline databases were systematically searched for relevant studies using related key terms and Medical Subject Headings (MeSH). Titles, abstracts, and full texts were screened, and data from eligible articles were extracted by two groups independently and in duplicate (two reviewers per group). Disagreements were resolved by a fifth independent reviewer. The included articles were all preclinical (translational) in vivo English studies that investigated and compared the efficacy profile between VSV-IFNβ and non-treatment controls in animal models. The risk of bias among the studies was assessed by two reviewers independently and in duplicate using SYRCLE’s risk-of-bias tool for animal studies; disparities were addressed by a third independent reviewer.ResultsAfter employing relevant MeSH and key terms, we identified 1598 articles. A total of 87 articles were either duplicates or conference proceedings and were thus excluded. Following title and abstract screening, 37 articles were included in the full-text assessment. Finally, 14 studies met the eligibility criteria. Forty-two experiments from the included studies examined the potential efficacy of VSV-IFNβ through different routes of administration, including intratumoral, intraperitoneal, and intravenous routes. Thirty-seven experiments reported positive outcomes. Meanwhile, five experiments reported negative outcomes, three and two of which examined intratumoral and intravenous VSV-IFNβ administration, respectively.ConclusionAlthough the majority of the included studies support the promising potential of VSV-IFNβ as an oncolytic virus, further research is necessary to ensure a safe and efficacious profile to translate its application into clinical trials.Systematic review registrationhttps://www.crd.york.ac.uk/PROSPERO/, identifier CRD42022335418.
Collapse
Affiliation(s)
- Abdulaziz Molham Moglan
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
| | - Omar A. Albaradie
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
| | - Fares Fayez Alsayegh
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
| | - Hussam Mohsen Alharbi
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
| | - Yahya Marwan Samman
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
| | - Mohammed M. Jalal
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Nizar H. Saeedi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Ahmad Bakur Mahmoud
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Taibah University, Almadinah Almunwarah, Saudi Arabia
- Strategic Research and Innovation Laboratories, Taibah University, Almadinah Almunwarah, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- *Correspondence: Ahmad Bakur Mahmoud, ; Almohanad A. Alkayyal,
| | - Almohanad A. Alkayyal
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- *Correspondence: Ahmad Bakur Mahmoud, ; Almohanad A. Alkayyal,
| |
Collapse
|
12
|
Zhang Y, Nagalo BM. Immunovirotherapy Based on Recombinant Vesicular Stomatitis Virus: Where Are We? Front Immunol 2022; 13:898631. [PMID: 35837384 PMCID: PMC9273848 DOI: 10.3389/fimmu.2022.898631] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/31/2022] [Indexed: 01/05/2023] Open
Abstract
Vesicular stomatitis virus (VSV), a negative-strand RNA virus of the Vesiculovirus genus, has demonstrated encouraging anti-neoplastic activity across multiple human cancer types. VSV is particularly attractive as an oncolytic agent because of its broad tropism, fast replication kinetics, and amenability to genetic manipulations. Furthermore, VSV-induced oncolysis can elicit a potent antitumor cytotoxic T-cell response to viral proteins and tumor-associated antigens, resulting in a long-lasting antitumor effect. Because of this multifaceted immunomodulatory property, VSV was investigated extensively as an immunovirotherapy alone or combined with other anticancer modalities, such as immune checkpoint blockade. Despite these recent opportunities to delineate synergistic and additive antitumor effects with existing anticancer therapies, FDA approval for the use of oncolytic VSV in humans has not yet been granted. This mini-review discusses factors that have prompted the use of VSV as an immunovirotherapy in human cancers and provides insights into future perspectives and research areas to improve VSV-based oncotherapy.
Collapse
Affiliation(s)
- Yuguo Zhang
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Bolni Marius Nagalo
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- *Correspondence: Bolni Marius Nagalo,
| |
Collapse
|
13
|
Brown M. Engaging Pattern Recognition Receptors in Solid Tumors to Generate Systemic Antitumor Immunity. Cancer Treat Res 2022; 183:91-129. [PMID: 35551657 DOI: 10.1007/978-3-030-96376-7_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Malignant tumors frequently exploit innate immunity to evade immune surveillance. The priming, function, and polarization of antitumor immunity fundamentally depends upon context provided by the innate immune system, particularly antigen presenting cells. Such context is determined in large part by sensing of pathogen specific and damage associated features by pathogen recognition receptors (PRRs). PRR activation induces the delivery of T cell priming cues (e.g. chemokines, co-stimulatory ligands, and cytokines) from antigen presenting cells, playing a decisive role in the cancer immunity cycle. Indeed, endogenous PRR activation within the tumor microenvironment (TME) has been shown to generate spontaneous antitumor T cell immunity, e.g., cGAS-STING mediated activation of antigen presenting cells after release of DNA from dying tumor cells. Thus, instigating intratumor PRR activation, particularly with the goal of generating Th1-promoting inflammation that stokes endogenous priming of antitumor CD8+ T cells, is a growing area of clinical investigation. This approach is analogous to in situ vaccination, ultimately providing a personalized antitumor response against relevant tumor associated antigens. Here I discuss clinical stage intratumor modalities that function via activation of PRRs. These approaches are being tested in various solid tumor contexts including melanoma, colorectal cancer, glioblastoma, head and neck squamous cell carcinoma, bladder cancer, and pancreatic cancer. Their mechanism (s) of action relative to other immunotherapy approaches (e.g., antigen-defined cancer vaccines, CAR T cells, dendritic cell vaccines, and immune checkpoint blockade), as well as their potential to complement these approaches are also discussed. Examples to be reviewed include TLR agonists, STING agonists, RIG-I agonists, and attenuated or engineered viruses and bacterium. I also review common key requirements for effective in situ immune activation, discuss differences between various strategies inclusive of mechanisms that may ultimately limit or preclude antitumor efficacy, and provide a summary of relevant clinical data.
Collapse
Affiliation(s)
- Michael Brown
- Department of Neurosurgery, Duke University, Durham, NC, USA.
| |
Collapse
|
14
|
Wang L, Chard Dunmall LS, Cheng Z, Wang Y. Remodeling the tumor microenvironment by oncolytic viruses: beyond oncolysis of tumor cells for cancer treatment. J Immunother Cancer 2022; 10:e004167. [PMID: 35640930 PMCID: PMC9157365 DOI: 10.1136/jitc-2021-004167] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2022] [Indexed: 12/25/2022] Open
Abstract
Tumor cells manipulate the local environment in which they grow, creating a tumor microenvironment (TME) that promotes tumor survival and metastasis. The TME is an extremely complex environment rich in immunosuppressive cells and cytokines. Various methods to therapeutically target the complicated TME are emerging as a potential approach for cancer treatment. Oncolytic viruses (OVs) are one of the most promising methods for remodeling the TME into an antitumor environment and can be used alone or in combination with other immunotherapy options. OVs replicate specifically in tumor cells and can be genetically engineered to target multiple elements of the TME simultaneously, thus representing a therapeutic with the potential to modify the TME to promote activation of antitumor immune cells and overcome tumor therapeutic resistance and recurrence. In this review, we analyze the tropism of OVs towards tumor cells and explore the interaction between OVs and immune cells, tumor stroma, vasculature and the metabolic environment in detail to help understand how OVs may be one of our most promising prospects for long-term curative therapies. We also discuss some of the challenges associated with TME therapies, and future perspectives in this evolving field.
Collapse
Affiliation(s)
- Lihong Wang
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention and Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Louisa S Chard Dunmall
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Zhenguo Cheng
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention and Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yaohe Wang
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention and Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, UK
| |
Collapse
|
15
|
Gourronc FA, Rebagliati M, Kramer-Riesberg B, Fleck AM, Patten JJ, Geohegan-Barek K, Messingham KN, Davey RA, Maury W, Klingelhutz AJ. Adipocytes are susceptible to Ebola Virus infection. Virology 2022; 573:12-22. [DOI: 10.1016/j.virol.2022.05.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 12/23/2022]
|
16
|
Evgin L, Kottke T, Tonne J, Thompson J, Huff AL, van Vloten J, Moore M, Michael J, Driscoll C, Pulido J, Swanson E, Kennedy R, Coffey M, Loghmani H, Sanchez-Perez L, Olivier G, Harrington K, Pandha H, Melcher A, Diaz RM, Vile RG. Oncolytic virus-mediated expansion of dual-specific CAR T cells improves efficacy against solid tumors in mice. Sci Transl Med 2022; 14:eabn2231. [PMID: 35417192 PMCID: PMC9297825 DOI: 10.1126/scitranslmed.abn2231] [Citation(s) in RCA: 82] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Oncolytic viruses (OVs) encoding a variety of transgenes have been evaluated as therapeutic tools to increase the efficacy of chimeric antigen receptor (CAR)-modified T cells in the solid tumor microenvironment (TME). Here, using systemically delivered OVs and CAR T cells in immunocompetent mouse models, we have defined a mechanism by which OVs can potentiate CAR T cell efficacy against solid tumor models of melanoma and glioma. We show that stimulation of the native T cell receptor (TCR) with viral or virally encoded epitopes gives rise to enhanced proliferation, CAR-directed antitumor function, and distinct memory phenotypes. In vivo expansion of dual-specific (DS) CAR T cells was leveraged by in vitro preloading with oncolytic vesicular stomatitis virus (VSV) or reovirus, allowing for a further in vivo expansion and reactivation of T cells by homologous boosting. This treatment led to prolonged survival of mice with subcutaneous melanoma and intracranial glioma tumors. Human CD19 CAR T cells could also be expanded in vitro with TCR reactivity against viral or virally encoded antigens and was associated with greater CAR-directed cytokine production. Our data highlight the utility of combining OV and CAR T cell therapy and show that stimulation of the native TCR can be exploited to enhance CAR T cell activity and efficacy in mice.
Collapse
Affiliation(s)
- Laura Evgin
- Department of Molecular Medicine, Mayo Clinic, Rochester,
MN 55905, USA
| | - Tim Kottke
- Department of Molecular Medicine, Mayo Clinic, Rochester,
MN 55905, USA
| | - Jason Tonne
- Department of Molecular Medicine, Mayo Clinic, Rochester,
MN 55905, USA
| | - Jill Thompson
- Department of Molecular Medicine, Mayo Clinic, Rochester,
MN 55905, USA
| | - Amanda L. Huff
- Department of Molecular Medicine, Mayo Clinic, Rochester,
MN 55905, USA
| | - Jacob van Vloten
- Department of Molecular Medicine, Mayo Clinic, Rochester,
MN 55905, USA
| | - Madelyn Moore
- Department of Molecular Medicine, Mayo Clinic, Rochester,
MN 55905, USA
| | - Josefine Michael
- Department of Molecular Medicine, Mayo Clinic, Rochester,
MN 55905, USA
| | | | - Jose Pulido
- Department of Molecular Medicine, Mayo Clinic, Rochester,
MN 55905, USA
| | - Eric Swanson
- Vaccine Research Group, Mayo Clinic, Rochester, MN 55905,
USA
| | - Richard Kennedy
- Vaccine Research Group, Mayo Clinic, Rochester, MN 55905,
USA
| | - Matt Coffey
- Oncolytics Biotech Incorporated, Calgary, AB, Canada
| | | | | | - Gloria Olivier
- Mayo Clinic Ventures, Mayo Clinic, Rochester, MN 55905,
USA
| | - Kevin Harrington
- Division of Radiotherapy and Imaging, Institute of Cancer
Research, Chester Beatty Laboratories, London SW3 6JB, UK
| | - Hardev Pandha
- Faculty of Health and Medical Sciences, University of
Surrey, Guildford GU2 7WG, UK
| | - Alan Melcher
- Division of Radiotherapy and Imaging, Institute of Cancer
Research, Chester Beatty Laboratories, London SW3 6JB, UK
| | - Rosa Maria Diaz
- Department of Molecular Medicine, Mayo Clinic, Rochester,
MN 55905, USA
| | - Richard G. Vile
- Department of Molecular Medicine, Mayo Clinic, Rochester,
MN 55905, USA
- Department of Immunology, Mayo Clinic, Rochester, MN 55905,
USA
| |
Collapse
|
17
|
Naumenko VA, Stepanenko AA, Lipatova AV, Vishnevskiy DA, Chekhonin VP. Infection of non-cancer cells: A barrier or support for oncolytic virotherapy? MOLECULAR THERAPY - ONCOLYTICS 2022; 24:663-682. [PMID: 35284629 PMCID: PMC8898763 DOI: 10.1016/j.omto.2022.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Oncolytic viruses are designed to specifically target cancer cells, sparing normal cells. Although numerous studies demonstrate the ability of oncolytic viruses to infect a wide range of non-tumor cells, the significance of this phenomenon for cancer virotherapy is poorly understood. To fill the gap, we summarize the data on infection of non-cancer targets by oncolytic viruses with a special focus on tumor microenvironment and secondary lymphoid tissues. The review aims to address two major questions: how do attenuated viruses manage to infect normal cells, and whether it is of importance for oncolytic virotherapy.
Collapse
Affiliation(s)
- Victor A. Naumenko
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow 119034, Russia
- Corresponding author Victor A. Naumenko, PhD, V. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow 119034, Russia.
| | - Aleksei A. Stepanenko
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow 119034, Russia
- Department of Medical Nanobiotechnology, N.I Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Anastasiia V. Lipatova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia
| | - Daniil A. Vishnevskiy
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow 119034, Russia
| | - Vladimir P. Chekhonin
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow 119034, Russia
- Department of Medical Nanobiotechnology, N.I Pirogov Russian National Research Medical University, Moscow 117997, Russia
| |
Collapse
|
18
|
Abd-Aziz N, Poh CL. Development of oncolytic viruses for cancer therapy. Transl Res 2021; 237:98-123. [PMID: 33905949 DOI: 10.1016/j.trsl.2021.04.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 02/07/2023]
Abstract
Oncolytic virotherapy is a therapeutic approach that uses replication-competent viruses to kill cancers. The ability of oncolytic viruses to selectively replicate in cancer cells leads to direct cell lysis and induction of anticancer immune response. Like other anticancer therapies, oncolytic virotherapy has several limitations such as viral delivery to the target, penetration into the tumor mass, and antiviral immune responses. This review provides an insight into the different characteristics of oncolytic viruses (natural and genetically modified) that contribute to effective applications of oncolytic virotherapy in preclinical and clinical trials, and strategies to overcome the limitations. The potential of oncolytic virotherapy combining with other conventional treatments or cancer immunotherapies involving immune checkpoint inhibitors and CAR-T therapy could form part of future multimodality treatment strategies.
Collapse
Affiliation(s)
- Noraini Abd-Aziz
- Centre for Virus and Vaccine Research (CVVR), School of Medical and Life Sciences, Sunway University, Subang Jaya, Selangor, Malaysia
| | - Chit Laa Poh
- Centre for Virus and Vaccine Research (CVVR), School of Medical and Life Sciences, Sunway University, Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
19
|
Cook J, Acosta-Medina AA, Peng KW, Lacy M, Russell S. Oncolytic virotherapy - Forging its place in the immunomodulatory paradigm for Multiple Myeloma. Cancer Treat Res Commun 2021; 29:100473. [PMID: 34673439 DOI: 10.1016/j.ctarc.2021.100473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 09/25/2021] [Indexed: 12/23/2022]
Abstract
The treatment focus for multiple myeloma (MM) has recently pivoted towards immune modulating strategies, with T-cell redirection therapies currently at the forefront of drug development. Yet, despite this revolution in treatment, MM remains without a sustainable cure. At the same time, tremendous advancement has been made in recombinant and gene editing techniques for oncolytic viruses (OV), which have increased their tumor specificity, improved safety, and enhanced the oncolytic and immunostimulatory potential. These breakthrough developments in oncolytic virotherapy have opened new avenues for OVs to be used in combination with other immune-based therapies such as checkpoint inhibitors, chimeric antigen receptor T-cells (CAR-T) and bispecific T-cell engagers. In this review, the authors place the spotlight on systemic oncolytic virotherapy as an adaptable immunotherapeutic for MM, highlight the unique mechanism of OVs in activating the immune-suppressive marrow microenvironment, and lastly showcase the OV platforms and the promising combination strategies in the pipeline for MM.
Collapse
Affiliation(s)
- Joselle Cook
- Division of Hematology, Mayo Clinic, Rochester MN, United States.
| | | | - Kah Whye Peng
- Department of Molecular Medicine, Mayo Clinic, Rochester MN , United States
| | - Martha Lacy
- Division of Hematology, Mayo Clinic, Rochester MN, United States
| | - Stephen Russell
- Division of Hematology, Mayo Clinic, Rochester MN, United States; Department of Molecular Medicine, Mayo Clinic, Rochester MN , United States
| |
Collapse
|
20
|
Mozaffari Nejad AS, Noor T, Munim ZH, Alikhani MY, Ghaemi A. A bibliometric review of oncolytic virus research as a novel approach for cancer therapy. Virol J 2021; 18:98. [PMID: 33980264 PMCID: PMC8113799 DOI: 10.1186/s12985-021-01571-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 05/03/2021] [Indexed: 02/06/2023] Open
Abstract
Background In recent years, oncolytic viruses (OVs) have drawn attention as a novel therapy to various types of cancers, both in clinical and preclinical cancer studies all around the world. Consequently, researchers have been actively working on enhancing cancer therapy since the early twentieth century. This study presents a systematic review of the literature on OVs, discusses underlying research clusters and, presents future directions of OVs research. Methods A total of 1626 published articles related to OVs as cancer therapy were obtained from the Web of Science (WoS) database published between January 2000 and March 2020. Various aspects of OVs research, including the countries/territories, institutions, journals, authors, citations, research areas, and content analysis to find trending and emerging topics, were analysed using the bibliometrix package in the R-software. Results In terms of the number of publications, the USA based researchers were the most productive (n = 611) followed by Chinese (n = 197), and Canadian (n = 153) researchers. The Molecular Therapy journal ranked first both in terms of the number of publications (n = 133) and local citations (n = 1384). The most prominent institution was Mayo Clinic from the USA (n = 117) followed by the University of Ottawa from Canada (n = 72), and the University of Helsinki from Finland (n = 63). The most impactful author was Bell J.C with the highest number of articles (n = 67) and total local citations (n = 885). The most impactful article was published in the Cell journal. In addition, the latest OVs research mainly builds on four research clusters. Conclusion The domain of OVs research has increased at a rapid rate from 2000 to 2020. Based on the synthesis of reviewed studies, adenovirus, herpes simplex virus, reovirus, and Newcastle disease virus have shown potent anti-cancer activity. Developed countries such as the USA, Canada, the UK, and Finland were the most productive, hence, contributed most to this field. Further collaboration will help improve the clinical research translation of this therapy and bring benefits to cancer patients worldwide.
Collapse
Affiliation(s)
| | - Tehjeeb Noor
- Faculty of Medicine, University of Bergen, Horten, Norway
| | - Ziaul Haque Munim
- Faculty of Technology, Natural and Maritime Sciences, University of South-Eastern Norway, Horten, Norway
| | - Mohammad Yousef Alikhani
- Department of Microbiology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Amir Ghaemi
- Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
21
|
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: 3.0] [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
|
22
|
Parking CAR T Cells in Tumours: Oncolytic Viruses as Valets or Vandals? Cancers (Basel) 2021; 13:cancers13051106. [PMID: 33807553 PMCID: PMC7961585 DOI: 10.3390/cancers13051106] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/01/2021] [Accepted: 03/03/2021] [Indexed: 12/18/2022] Open
Abstract
Oncolytic viruses (OVs) and adoptive T cell therapy (ACT) each possess direct tumour cytolytic capabilities, and their combination potentially seems like a match made in heaven to complement the strengths and weakness of each modality. While providing strong innate immune stimulation that can mobilize adaptive responses, the magnitude of anti-tumour T cell priming induced by OVs is often modest. Chimeric antigen receptor (CAR) modified T cells bypass conventional T cell education through introduction of a synthetic receptor; however, realization of their full therapeutic properties can be stunted by the heavily immune-suppressive nature of the tumour microenvironment (TME). Oncolytic viruses have thus been seen as a natural ally to overcome immunosuppressive mechanisms in the TME which limit CAR T cell infiltration and functionality. Engineering has further endowed viruses with the ability to express transgenes in situ to relieve T cell tumour-intrinsic resistance mechanisms and decorate the tumour with antigen to overcome antigen heterogeneity or loss. Despite this helpful remodeling of the tumour microenvironment, it has simultaneously become clear that not all virus induced effects are favourable for CAR T, begging the question whether viruses act as valets ushering CAR T into their active site, or vandals which cause chaos leading to both tumour and T cell death. Herein, we summarize recent studies combining these two therapeutic modalities and seek to place them within the broader context of viral T cell immunology which will help to overcome the current limitations of effective CAR T therapy to make the most of combinatorial strategies.
Collapse
|
23
|
Askari FS, Mohebbi A, Moradi A, Javid N. The Role of Vesicular Stomatitis Virus Matrix Protein in Autophagy in the Breast Cancer. Asian Pac J Cancer Prev 2021; 22:249-255. [PMID: 33507706 PMCID: PMC8184201 DOI: 10.31557/apjcp.2021.22.1.249] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Breast cancer is one of the most difficult malignancies to treat. Therapeutics is used to target and kill the cancer cells. Non-human oncolytic viruses have the ability to cause cell death directly to cancers. The objective here was to investigate the role of Vesicular Stomatitis Virus (VSV) Matrix (M) protein in autophagy in the breast cancer cell line. METHODS Two different VSV wild type and mutant (M51R) M protein constructs were produced. Breast cancer cell line BT-20 was transfected by either wild type or mutant vectors. Transfection efficiency was measured using a fluorescent microscopy. Expression of VSV M protein was investigated at protein level. Cell cytotoxicity was measured using an MTT assay. The autophagy pathway was studied by Beclin-1 immunoassay. Data were statistically analyzed between different transfected groups. RESULTS It has been shown that the VSV M protein induced higher levels of Beclin-1 than the M51R mutant in the BT-20 cell line. Increased levels of Beclin-1 were also associated with VSV M cell-induced cytotoxicity. CONCLUSION It has been shown here that VSV wild type or mutant M proteins can cause autophagy-induced cell death by increasing Beclin-1 expression. This includes the possible role of VSV to be used as an oncolytic virus in breast cancer treatment. <br />.
Collapse
Affiliation(s)
- Fatemeh Sana Askari
- Student Research Committee, School of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Alireza Mohebbi
- Stem Cell Research Center, School of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Abdolvahab Moradi
- Department of Microbiology, School of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Naeme Javid
- Department of Microbiology, School of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
24
|
Tell JG, Coller BAG, Dubey SA, Jenal U, Lapps W, Wang L, Wolf J. Environmental Risk Assessment for rVSVΔG-ZEBOV-GP, a Genetically Modified Live Vaccine for Ebola Virus Disease. Vaccines (Basel) 2020; 8:vaccines8040779. [PMID: 33352786 PMCID: PMC7767225 DOI: 10.3390/vaccines8040779] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 12/17/2020] [Accepted: 12/17/2020] [Indexed: 01/04/2023] Open
Abstract
rVSVΔG-ZEBOV-GP is a live, attenuated, recombinant vesicular stomatitis virus (rVSV)-based vaccine for the prevention of Ebola virus disease caused by Zaire ebolavirus. As a replication-competent genetically modified organism, rVSVΔG-ZEBOV-GP underwent various environmental evaluations prior to approval, the most in-depth being the environmental risk assessment (ERA) required by the European Medicines Agency. This ERA, as well as the underlying methodology used to arrive at a sound conclusion about the environmental risks of rVSVΔG-ZEBOV-GP, are described in this review. Clinical data from vaccinated adults demonstrated only infrequent, low-level shedding and transient, low-level viremia, indicating a low person-to-person infection risk. Animal data suggest that it is highly unlikely that vaccinated individuals would infect animals with recombinant virus vaccine or that rVSVΔG-ZEBOV-GP would spread within animal populations. Preclinical studies in various hematophagous insect vectors showed that these species were unable to transmit rVSVΔG-ZEBOV-GP. Pathogenicity risk in humans and animals was found to be low, based on clinical and preclinical data. The overall risk for non-vaccinated individuals and the environment is thus negligible and can be minimized further through defined mitigation strategies. This ERA and the experience gained are relevant to developing other rVSV-based vaccines, including candidates under investigation for prevention of COVID-19.
Collapse
Affiliation(s)
- Joan G. Tell
- Merck & Co., Inc., Kenilworth, NJ 07033, USA; (B.-A.G.C.); (S.A.D.); (W.L.); (L.W.); (J.W.)
- Correspondence:
| | - Beth-Ann G. Coller
- Merck & Co., Inc., Kenilworth, NJ 07033, USA; (B.-A.G.C.); (S.A.D.); (W.L.); (L.W.); (J.W.)
| | - Sheri A. Dubey
- Merck & Co., Inc., Kenilworth, NJ 07033, USA; (B.-A.G.C.); (S.A.D.); (W.L.); (L.W.); (J.W.)
| | - Ursula Jenal
- Jenal & Partners Biosafety Consulting, 4310 Rheinfelden, Switzerland;
| | - William Lapps
- Merck & Co., Inc., Kenilworth, NJ 07033, USA; (B.-A.G.C.); (S.A.D.); (W.L.); (L.W.); (J.W.)
| | - Liman Wang
- Merck & Co., Inc., Kenilworth, NJ 07033, USA; (B.-A.G.C.); (S.A.D.); (W.L.); (L.W.); (J.W.)
| | - Jayanthi Wolf
- Merck & Co., Inc., Kenilworth, NJ 07033, USA; (B.-A.G.C.); (S.A.D.); (W.L.); (L.W.); (J.W.)
| |
Collapse
|
25
|
Jamieson TR, Poutou J, Ilkow CS. Redirecting oncolytic viruses: Engineering opportunists to take control of the tumour microenvironment. Cytokine Growth Factor Rev 2020; 56:102-114. [DOI: 10.1016/j.cytogfr.2020.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 07/02/2020] [Indexed: 12/12/2022]
|
26
|
Hamada M, Yura Y. Efficient Delivery and Replication of Oncolytic Virus for Successful Treatment of Head and Neck Cancer. Int J Mol Sci 2020; 21:E7073. [PMID: 32992948 PMCID: PMC7582277 DOI: 10.3390/ijms21197073] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/18/2020] [Accepted: 09/22/2020] [Indexed: 12/14/2022] Open
Abstract
Head and neck cancer has been treated by a combination of surgery, radiation, and chemotherapy. In recent years, the development of immune checkpoint inhibitors (ICIs) has made immunotherapy a new treatment method. Oncolytic virus (OV) therapy selectively infects tumor cells with a low-pathogenic virus, lyses tumor cells by the cytopathic effects of the virus, and induces anti-tumor immunity to destroy tumors by the action of immune cells. In OV therapy for head and neck squamous cell carcinoma (HNSCC), viruses, such as herpes simplex virus type 1 (HSV-1), vaccinia virus, adenovirus, reovirus, measles virus, and vesicular stomatitis virus (VSV), are mainly used. As the combined use of mutant HSV-1 and ICI was successful for the treatment of melanoma, studies are underway to combine OV therapy with radiation, chemotherapy, and other types of immunotherapy. In such therapy, it is important for the virus to selectively replicate in tumor cells, and to express the viral gene and the introduced foreign gene in the tumor cells. In OV therapy for HNSCC, it may be useful to combine systemic and local treatments that improve the delivery and replication of the inoculated oncolytic virus in the tumor cells.
Collapse
Affiliation(s)
- Masakazu Hamada
- Department of Oral and Maxillofacial Surgery, Osaka University Graduate School of Dentistry, Suita, Osaka 565-0871, Japan;
| | | |
Collapse
|
27
|
Munis AM, Bentley EM, Takeuchi Y. A tool with many applications: vesicular stomatitis virus in research and medicine. Expert Opin Biol Ther 2020; 20:1187-1201. [PMID: 32602788 DOI: 10.1080/14712598.2020.1787981] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Vesicular stomatitis virus (VSV) has long been a useful research tool in virology and recently become an essential part of medicinal products. Vesiculovirus research is growing quickly following its adaptation to clinical gene and cell therapy and oncolytic virotherapy. AREAS COVERED This article reviews the versatility of VSV as a research tool and biological reagent, its use as a viral and vaccine vector delivering therapeutic and immunogenic transgenes and an oncolytic virus aiding cancer treatment. Challenges such as the immune response against such advanced therapeutic medicinal products and manufacturing constraints are also discussed. EXPERT OPINION The field of in vivo gene and cell therapy is advancing rapidly with VSV used in many ways. Comparison of VSV's use as a versatile therapeutic reagent unveils further prospects and problems for each application. Overcoming immunological challenges to aid repeated administration of viral vectors and minimizing harmful host-vector interactions remains one of the major challenges. In the future, exploitation of reverse genetic tools may assist the creation of recombinant viral variants that have improved onco-selectivity and more efficient vaccine vector activity. This will add to the preferential features of VSV as an excellent advanced therapy medicinal product (ATMP) platform.
Collapse
Affiliation(s)
- Altar M Munis
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford , Oxford, UK.,Division of Advanced Therapies, National Institute for Biological Standards and Control , South Mimms, UK
| | - Emma M Bentley
- Division of Virology, National Institute for Biological Standards and Control , South Mimms, UK
| | - Yasuhiro Takeuchi
- Division of Advanced Therapies, National Institute for Biological Standards and Control , South Mimms, UK.,Division of Infection and Immunity, University College London , London, UK
| |
Collapse
|
28
|
Faria Waziry PA, Raja A, Salmon C, Aldana N, Damodar S, Fukushima AR, Mayi BS. Impact of pyriproxyfen on virus behavior: implications for pesticide-induced virulence and mechanism of transmission. Virol J 2020; 17:93. [PMID: 32631404 PMCID: PMC7339562 DOI: 10.1186/s12985-020-01378-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 06/30/2020] [Indexed: 02/06/2023] Open
Abstract
Background More than 3 years since the last Zika virus (ZIKV) outbreak in Brazil, researchers are still deciphering the molecular mechanisms of neurovirulence and vertical transmission, as well as the best way to control spread of ZIKV, a flavivirus. The use of pesticides was the main strategy of mosquito control during the last ZIKV outbreak. Methods We used vesicular stomatitis virus (VSV) tagged with green fluorescent protein (GFP) as our prototypical virus to study the impact of insecticide pyriproxyfen (PPF). VZV-GFP infected and uninfected Jurkat, HeLa and trophoblast cells were treated with PPF and compared to untreated cells (control). Cell viability was determined by the MTT assay. Cell morphology, presence of extracellular vesicles (EVs), virus infection/GFP expression as well as active mitochondrial levels/localization were examined by confocal microscopy. Results PPF, which was used to control mosquito populations in Brazil prior to the ZIKV outbreak, enhances VSV replication and has cell membrane-altering properties in the presence of virus. PPF causes enhanced viral replication and formation of large EVs, loaded with virus as well as mitochondria. Treatment of trophoblasts or HeLa cells with increasing concentrations of PPF does not alter cell viability, however, it proportionately increases Jurkat cell viability. Increasing concentrations of PPF followed by VSV infection does not interfere with HeLa cell viability. Both Jurkats and trophoblasts show proportionately increased cell death with increased concentrations of PPF in the presence of virus. Conclusions We hypothesize that PPF disrupts the lipid microenvironment of mammalian cells, thereby interfering with pathways of viral replication. PPF lowers viability of trophoblasts and Jurkats in the presence of VSV, implying that the combination renders immune system impairment in infected individuals as well as enhanced vulnerability of fetuses towards viral vertical transmission. We hypothesize that similar viruses such as ZIKV may be vertically transmitted via EV-to-cell contact when exposed to PPF, thereby bypassing immune detection. The impact of pesticides on viral replication must be fully investigated before large scale use in future outbreaks of mosquito borne viruses.
Collapse
Affiliation(s)
- Paula A Faria Waziry
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, 3400 Gulf to Bay Blvd, Clearwater, FL, 33759, USA
| | - Aarti Raja
- Department of Biological Sciences, Halmos College of Arts and Sciences, Nova Southeastern University, Fort Lauderdale, FL, 33314, USA
| | - Chloe Salmon
- Plymouth University, 3 Endsleigh Place, Drake Circus, Plymouth, England, PL4 8AA
| | - Nathalia Aldana
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, 3200 S. University Dr, Fort Lauderdale, FL, 33328, USA
| | - Sruthi Damodar
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, 3200 S. University Dr, Fort Lauderdale, FL, 33328, USA
| | - Andre Rinaldi Fukushima
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of Sao Paulo, São Paulo, Brazil
| | - Bindu S Mayi
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, 3400 Gulf to Bay Blvd, Clearwater, FL, 33759, USA.
| |
Collapse
|
29
|
Evgin L, Huff AL, Wongthida P, Thompson J, Kottke T, Tonne J, Schuelke M, Ayasoufi K, Driscoll CB, Shim KG, Reynolds P, Monie DD, Johnson AJ, Coffey M, Young SL, Archer G, Sampson J, Pulido J, Perez LS, Vile R. Oncolytic virus-derived type I interferon restricts CAR T cell therapy. Nat Commun 2020; 11:3187. [PMID: 32581235 PMCID: PMC7314766 DOI: 10.1038/s41467-020-17011-z] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 05/29/2020] [Indexed: 01/14/2023] Open
Abstract
The application of adoptive T cell therapies, including those using chimeric antigen receptor (CAR)-modified T cells, to solid tumors requires combinatorial strategies to overcome immune suppression associated with the tumor microenvironment. Here we test whether the inflammatory nature of oncolytic viruses and their ability to remodel the tumor microenvironment may help to recruit and potentiate the functionality of CAR T cells. Contrary to our hypothesis, VSVmIFNβ infection is associated with attrition of murine EGFRvIII CAR T cells in a B16EGFRvIII model, despite inducing a robust proinflammatory shift in the chemokine profile. Mechanistically, type I interferon (IFN) expressed following infection promotes apoptosis, activation, and inhibitory receptor expression, and interferon-insensitive CAR T cells enable combinatorial therapy with VSVmIFNβ. Our study uncovers an unexpected mechanism of therapeutic interference, and prompts further investigation into the interaction between CAR T cells and oncolytic viruses to optimize combination therapy.
Collapse
MESH Headings
- Animals
- Apoptosis
- Cell Line, Tumor
- Chemokines/metabolism
- Combined Modality Therapy
- Female
- Immunotherapy, Adoptive
- Interferon-beta/genetics
- Interferon-beta/metabolism
- Lymphocyte Activation
- Melanoma, Experimental/immunology
- Melanoma, Experimental/therapy
- Mice
- Mice, Inbred C57BL
- Mice, Mutant Strains
- Oncolytic Virotherapy
- Oncolytic Viruses/genetics
- Oncolytic Viruses/metabolism
- Receptor, Interferon alpha-beta/genetics
- Receptor, Interferon alpha-beta/metabolism
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Chimeric Antigen/metabolism
- Spleen/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Laura Evgin
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Amanda L Huff
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Jill Thompson
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Tim Kottke
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jason Tonne
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Kevin G Shim
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | - Pierce Reynolds
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Dileep D Monie
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Matt Coffey
- Oncolytics Biotech Incorporated, Calgary, Canada
| | - Sarah L Young
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Gary Archer
- Department of Neurosurgery, Duke University, Durham, NC, USA
| | - John Sampson
- Department of Neurosurgery, Duke University, Durham, NC, USA
| | - Jose Pulido
- Department of Ophthalmology, Mayo Clinic, Rochester, MN, USA
| | | | - Richard Vile
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA.
- Department of Immunology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
30
|
Bayne RS, Puckett S, Rodrigues LU, Cramer SD, Lee J, Furdui CM, Chou JW, Miller LD, Ornelles DA, Lyles DS. MAP3K7 and CHD1 Are Novel Mediators of Resistance to Oncolytic Vesicular Stomatitis Virus in Prostate Cancer Cells. MOLECULAR THERAPY-ONCOLYTICS 2020; 17:496-507. [PMID: 32529027 PMCID: PMC7276393 DOI: 10.1016/j.omto.2020.05.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 05/14/2020] [Indexed: 12/15/2022]
Abstract
A key principle of oncolytic viral therapy is that many cancers develop defects in their antiviral responses, making them more susceptible to virus infection. However, some cancers display resistance to viral infection. Many of these resistant cancers constitutively express interferon-stimulated genes (ISGs). The goal of these experiments was to determine the role of two tumor suppressor genes, MAP3K7 and CHD1, in viral resistance and ISG expression in PC3 prostate cancer cells resistant to oncolytic vesicular stomatitis virus (VSV). MAP3K7 and CHD1 are often co-deleted in aggressive prostate cancers. Silencing expression of MAP3K7 and CHD1 in PC3 cells increased susceptibility to the matrix (M) gene mutant M51R-VSV, as shown by increased expression of viral genes, increased yield of progeny virus, and reduction of tumor growth in nude mice. Silencing MAP3K7 alone had a greater effect on virus susceptibility than did silencing CHD1. Silencing MAP3K7 and CHD1 decreased constitutive expression of ISG mRNAs and proteins, whereas silencing MAP3K7 alone decreased expression of ISG proteins, but actually increased expression of ISG mRNAs. These results suggest a role for the protein product of MAP3K7, transforming growth factor β-activated kinase 1 (TAK1), in regulating translation of ISG mRNAs and a role of CHD1 in maintaining the transcription of ISGs.
Collapse
Affiliation(s)
- Robert S Bayne
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Shelby Puckett
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | | - Scott D Cramer
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jingyun Lee
- Department of Internal Medicine, Section on Molecular Medicine, Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Cristina M Furdui
- Department of Internal Medicine, Section on Molecular Medicine, Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jeff W Chou
- Department of Biostatistical Sciences, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Lance D Miller
- 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
| | - Douglas S Lyles
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
31
|
Udayakumar TS, Betancourt DM, Ahmad A, Tao W, Totiger TM, Patel M, Marples B, Barber G, Pollack A. Radiation Attenuates Prostate Tumor Antiviral Responses to Vesicular Stomatitis Virus Containing IFNβ, Resulting in Pronounced Antitumor Systemic Immune Responses. Mol Cancer Res 2020; 18:1232-1243. [PMID: 32366674 DOI: 10.1158/1541-7786.mcr-19-0836] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 11/26/2019] [Accepted: 04/30/2020] [Indexed: 11/16/2022]
Abstract
Vesicular stomatitis virus (VSV) expressing IFNβ induces apoptosis in multiple tumor models while maintaining an excellent safety profile. VSV-IFNβ is oncoselective due to permissive replication in cells with an altered IFN pathway. The human VSV-IFNβ (hIFNβ) vector is currently used in clinical trials as a standalone therapy; however, we hypothesized that oncolytic virotherapy might be more effective when used in combination with radiotherapy (RT). We investigated the synergistic effects of RT and VSV-hIFNβ in the subcutaneous PC3 and orthotopic LNCaP prostate xenograft models and a syngeneic RM9 prostate tumor model. VSV-IFNβ combined with RT amplified tumor killing for PC3 and LNCaP xenografts, and RM9 tumors. This was attributed to the induction of proapoptotic genes leading to increased VSV-IFNβ infection and replication, VSV expression, and oncolysis. In the RM9 tumors, combination therapy resulted in a robust antitumor immune response. Treated RM9 tumor-bearing mice demonstrated an increase in CD8+ and CD4+ T-cell numbers, 100% resistance to tumor rechallenge, and reduced resistance to reimplantation challenge with CD8+ knockdown. RT enhanced the activity of VSV-mediated oncolysis via attenuation of the innate antiviral response, resulting in increased VSV replication and the generation of an adaptive immune response earmarked by an increase in CD8+ lymphocyte numbers and antitumor activity. Local tumor irradiation combined with VSV-IFNβ affects tumor cell death through direct and systemic activity in conjunction with pronounced antitumor immunity. IMPLICATIONS: Radiotherapy enhances VSV-mediated oncolysis and anti-tumor immunity, indicating that the ombination has promise for very high risk prostate cancer.
Collapse
Affiliation(s)
- Thirupandiyur S Udayakumar
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Dillon M Betancourt
- Department of Cell Biology, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Anis Ahmad
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Wensi Tao
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Tulasigeri M Totiger
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Mausam Patel
- Department of Radiology, Memorial Health, Savannah, Georgia
| | - Brian Marples
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Glen Barber
- Department of Cell Biology, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Alan Pollack
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida.
| |
Collapse
|
32
|
Gélinas JF, Azizi H, Kiesslich S, Lanthier S, Perdersen J, Chahal PS, Ansorge S, Kobinger G, Gilbert R, Kamen AA. Production of rVSV-ZEBOV in serum-free suspension culture of HEK 293SF cells. Vaccine 2019; 37:6624-6632. [DOI: 10.1016/j.vaccine.2019.09.044] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/28/2019] [Accepted: 09/11/2019] [Indexed: 12/13/2022]
|
33
|
Ogino M, Fedorov Y, Adams DJ, Okada K, Ito N, Sugiyama M, Ogino T. Vesiculopolins, a New Class of Anti-Vesiculoviral Compounds, Inhibit Transcription Initiation of Vesiculoviruses. Viruses 2019; 11:v11090856. [PMID: 31540123 PMCID: PMC6783830 DOI: 10.3390/v11090856] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 09/11/2019] [Accepted: 09/11/2019] [Indexed: 01/09/2023] Open
Abstract
Vesicular stomatitis virus (VSV) represents a promising platform for developing oncolytic viruses, as well as vaccines against significant human pathogens. To safely control VSV infection in humans, small-molecule drugs that selectively inhibit VSV infection may be needed. Here, using a cell-based high-throughput screening assay followed by an in vitro transcription assay, compounds with a 7-hydroxy-6-methyl-3,4-dihydroquinolin-2(1H)-one structure and an aromatic group at position 4 (named vesiculopolins, VPIs) were identified as VSV RNA polymerase inhibitors. The most effective compound, VPI A, inhibited VSV-induced cytopathic effects and in vitro mRNA synthesis with micromolar to submicromolar 50% inhibitory concentrations. VPI A was found to inhibit terminal de novo initiation rather than elongation for leader RNA synthesis, but not mRNA capping, with the VSV L protein, suggesting that VPI A is targeted to the polymerase domain in the L protein. VPI A inhibited transcription of Chandipura virus, but not of human parainfluenza virus 3, suggesting that it specifically acts on vesiculoviral L proteins. These results suggest that VPIs may serve not only as molecular probes to elucidate the mechanisms of transcription of vesiculoviruses, but also as lead compounds to develop antiviral drugs against vesiculoviruses and other related rhabdoviruses.
Collapse
Affiliation(s)
- Minako Ogino
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | - Yuriy Fedorov
- Small Molecule Drug Development Core, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | - Drew J Adams
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | - Kazuma Okada
- Laboratory of Zoonotic Diseases, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan.
| | - Naoto Ito
- Laboratory of Zoonotic Diseases, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan.
- Gifu Center for Highly Advanced Integration of Nanosciences and Life Sciences (G-CHAIN), Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan.
| | - Makoto Sugiyama
- Laboratory of Zoonotic Diseases, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan.
| | - Tomoaki Ogino
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
34
|
Sakuda T, Kubo T, Johan MP, Furuta T, Sakaguchi T, Nakanishi M, Ochi M, Adachi N. Novel Near-Infrared Fluorescence-Guided Surgery With Vesicular Stomatitis Virus for Complete Surgical Resection of Osteosarcomas in Mice. J Orthop Res 2019; 37:1192-1201. [PMID: 30839125 DOI: 10.1002/jor.24277] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 02/20/2019] [Indexed: 02/04/2023]
Abstract
Attempts have been made to visualize tumor cells intraoperatively with fluorescence guidance. However, the clear demarcation and complete tumor resection have always been a challenging task. To address this, we have developed a novel fluorescence bioimaging system with vesicular stomatitis virus (VSV) incorporating Katushka, near-infrared fluorescent protein. VSV is tumor-specific owing to the deficiency of antiviral interferon signaling pathways in tumor cells. We aimed to evaluate the tumor specificity of the recombinant VSV-Katushka (rVSV-K) in osteosarcoma cells and to assess the feasibility of complete tumor resection by the rVSV-K fluorescence guidance. In in vitro experiments, mouse and human osteosarcoma cell lines and normal human mesenchymal stem cells were infected with rVSV-K and observed by fluorescence microscopy. Near-infrared fluorescence was observed only in osteosarcoma cells, even at a low-concentration of virus infections. In in vivo experiments, mouse osteosarcoma (LM8) cells were transplanted subcutaneously into the back of immune-competent mice to produce an osteosarcoma, which was then injected with rVSV-K. The areas emitting fluorescence were resected using a bioimaging system. The distance between the surgical and tumor margins of the fluorescence-guided resection with rVSV-K group was significantly larger than that of the non-guided resection groups. The local recurrence rate was significantly lower in the fluorescence-guided resection with rVSV-K group than in the non-guided resection groups. The distant metastasis rate and average survival rate were not significantly different between all groups. These results suggest that the rVSV-K is specific to osteosarcoma cells and enables complete tumor resection of osteosarcomas in mice. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.
Collapse
Affiliation(s)
- Tomohiko Sakuda
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Tadahiko Kubo
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Muhammad Phetrus Johan
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.,Department of Orthopedic and Traumatology, Faculty of Medicine, Hasanuddin University, Tamalanrea Makassar, Indonesia
| | - Taisuke Furuta
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Takemasa Sakaguchi
- Department of Virology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Mahito Nakanishi
- National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Mitsuo Ochi
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Nobuo Adachi
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| |
Collapse
|
35
|
Gray Z, Tabarraei A, Moradi A, Kalani MR. M51R and Delta-M51 matrix protein of the vesicular stomatitis virus induce apoptosis in colorectal cancer cells. Mol Biol Rep 2019; 46:3371-3379. [PMID: 31006094 DOI: 10.1007/s11033-019-04799-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 04/05/2019] [Indexed: 12/17/2022]
Abstract
Colorectal cancer (CRC) is the third most common cancer in both men and women. Oncolytic viral-based therapy methods seem to be promising for CRC treatment. Vesicular stomatitis virus (VSV) is considered as a potent candidate in viral therapy for several tumors. VSV particles with mutated matrix (M) protein are capable of initiating cell death cascades while not being harmful to the immune system. In the current study, the effects of the VSV M-protein was investigated on the apoptosis of the colorectal cancer SW480 cell. Wild-type, M51R, and ΔM51 mutants VSV M-protein genes were cloned into the PCDNA3.1 vector and transfected into the SW480 cells. The results of the MTT assay, Western blotting, and Caspase 3, 8, and 9 measurement, illustrated that both wild and M51R mutant M-proteins can destroy the SW480 colorectal cancer cells. DAPI/TUNEL double-staining reconfirmed the apoptotic effects of the M-protein expression. The ΔM51 mutant M-protein is effective likewise M51R, somehow it can be considered as a safer substitution.
Collapse
Affiliation(s)
- Zahra Gray
- Department of Microbiology, College of Medicine, Golestan University of Medical Science, 1 Shastcola Ave, 5 km Sari Rd, Gorgan, Iran
| | - Alijan Tabarraei
- Department of Microbiology, College of Medicine, Golestan University of Medical Science, 1 Shastcola Ave, 5 km Sari Rd, Gorgan, Iran
| | - Abdolvahab Moradi
- Department of Microbiology, College of Medicine, Golestan University of Medical Science, 1 Shastcola Ave, 5 km Sari Rd, Gorgan, Iran.
| | - Mohamad R Kalani
- Cell and Molecular Research center, Golestan University of Medical Science, 1 Shastcola Ave, 5 km Sari Rd, Gorgan, Iran. .,Molecular and Cell Biology, RAL, University of Illinois at Urbana-Champaign, 600 S Goodwin Ave #325, Urbana, IL, 61801, USA.
| |
Collapse
|
36
|
Huff AL, Wongthida P, Kottke T, Thompson JM, Driscoll CB, Schuelke M, Shim KG, Harris RS, Molan A, Pulido JS, Selby PJ, Harrington KJ, Melcher A, Evgin L, Vile RG. APOBEC3 Mediates Resistance to Oncolytic Viral Therapy. Mol Ther Oncolytics 2018; 11:1-13. [PMID: 30294666 PMCID: PMC6169432 DOI: 10.1016/j.omto.2018.08.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/23/2018] [Indexed: 12/18/2022] Open
Abstract
Tumor cells frequently evade applied therapies through the accumulation of genomic mutations and rapid evolution. In the case of oncolytic virotherapy, understanding the mechanisms by which cancer cells develop resistance to infection and lysis is critical to the development of more effective viral-based platforms. Here, we identify APOBEC3 as an important factor that restricts the potency of oncolytic vesicular stomatitis virus (VSV). We show that VSV infection of B16 murine melanoma cells upregulated APOBEC3 in an IFN-β-dependent manner, which was responsible for the evolution of virus-resistant cell populations and suggested that APOBEC3 expression promoted the acquisition of a virus-resistant phenotype. Knockdown of APOBEC3 in B16 cells diminished their capacity to develop resistance to VSV infection in vitro and enhanced the therapeutic effect of VSV in vivo. Similarly, overexpression of human APOBEC3B promoted the acquisition of resistance to oncolytic VSV both in vitro and in vivo. Finally, we demonstrate that APOBEC3B expression had a direct effect on the fitness of VSV, an RNA virus that has not previously been identified as restricted by APOBEC3B. This research identifies APOBEC3 enzymes as key players to target in order to improve the efficacy of viral or broader nucleic acid-based therapeutic platforms.
Collapse
Affiliation(s)
- Amanda L. Huff
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Timothy Kottke
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Jill M. Thompson
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | - Kevin G. Shim
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Reuben S. Harris
- Howard Hughes Medical Institute, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Institute for Molecular Virology, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Amy Molan
- Howard Hughes Medical Institute, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Institute for Molecular Virology, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jose S. Pulido
- Department of Ophthalmology, Mayo Clinic, Rochester, MN 55905, USA
| | - Peter J. Selby
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James’s University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | | | | | - Laura Evgin
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Richard G. Vile
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James’s University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| |
Collapse
|
37
|
Nguyen NV, Tran JT, Sanchez DJ. HIV blocks Type I IFN signaling through disruption of STAT1 phosphorylation. Innate Immun 2018; 24:490-500. [PMID: 30282499 PMCID: PMC6513668 DOI: 10.1177/1753425918803674] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 08/30/2018] [Accepted: 09/05/2018] [Indexed: 11/26/2022] Open
Abstract
This study investigates the modulation of Type I IFN induction of an antiviral state by HIV. IFNs, including IFN-α, are key innate immune cytokines that activate the JAK/STAT pathway leading to the expression of IFN-stimulated genes. IFN-stimulated gene expression establishes the antiviral state, limiting viral infection in IFN-α-stimulated microenvironments. Our previous studies have shown that HIV proteins disrupt the induction of IFN-α by degradation of IFN-β promoter stimulator-1, an adaptor protein for the up-regulation and release of IFN-α into the local microenvironment via the retinoic acid-inducible gene 1-like receptor signaling pathway. However, IFN-α is still released from other sources such as plasmacytoid dendritic cells via TLR-dependent recognition of HIV. Here we report that the activation of the JAK/STAT pathway by IFN-α stimulation is disrupted by HIV proteins Vpu and Nef, which both reduce IFN-α induction of STAT1 phosphorylation. Thus, HIV would still be able to avoid antiviral protection induced by IFN-α in the local microenvironment. These findings show that HIV blocks multiple signaling points that would lead to the up-regulation of IFN-stimulated genes, allowing more effective replication in IFN-α-rich environments.
Collapse
Affiliation(s)
- Nam V Nguyen
- Department of Pharmaceutical Sciences, Western
University of Health Sciences, United States
| | - James T Tran
- Department of Pharmaceutical Sciences, Western
University of Health Sciences, United States
| | - David Jesse Sanchez
- Department of Pharmaceutical Sciences, Western
University of Health Sciences, United States
| |
Collapse
|
38
|
Russell L, Peng KW. The emerging role of oncolytic virus therapy against cancer. Chin Clin Oncol 2018; 7:16. [PMID: 29764161 DOI: 10.21037/cco.2018.04.04] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 04/10/2018] [Indexed: 12/28/2022]
Abstract
This review discusses current clinical advancements in oncolytic viral therapy, with a focus on the viral platforms approved for clinical use and highlights the benefits each platform provides. Three oncolytic viruses (OVs), an echovirus, an adenovirus, and a herpes simplex-1 virus, have passed governmental regulatory approval in Latvia, China, and the USA and EU. Numerous other recombinant viruses from diverse families are in clinical testing in cancer patients and we highlight the design features of selected examples, including adenovirus, herpes simplex virus, measles virus, retrovirus, reovirus, vaccinia virus, vesicular stomatitis virus. Lastly, we provide thoughts on the path forward for this rapidly expanding field especially in combination with immune modulating drugs.
Collapse
|
39
|
Zhang X, Mao G, van den Pol AN. Chikungunya-vesicular stomatitis chimeric virus targets and eliminates brain tumors. Virology 2018; 522:244-259. [PMID: 30055515 DOI: 10.1016/j.virol.2018.06.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/26/2018] [Accepted: 06/28/2018] [Indexed: 01/17/2023]
Abstract
Vesicular stomatitis virus (VSV) shows potential for targeting and killing cancer cells, but can be dangerous in the brain due to its neurotropic glycoprotein. Here we test a chimeric virus in which the VSV glycoprotein is replaced with the Chikungunya polyprotein E3-E2-6K-E1 (VSVΔG-CHIKV). Control mice with brain tumors survived a mean of 40 days after tumor implant. VSVΔG-CHIKV selectively infected and eliminated the tumor, and extended survival substantially in all tumor-bearing mice to over 100 days. VSVΔG-CHIKV also targeted intracranial primary patient derived melanoma xenografts. Virus injected into one melanoma spread to other melanomas within the same brain with little detectable infection of normal cells. Intravenous VSVΔG-CHIKV infected tumor cells but not normal tissue. In immunocompetent mice, VSVΔG-CHIKV selectively infected mouse melanoma cells within the brain. These data suggest VSVΔG-CHIKV can target and destroy brain tumors in multiple animal models without the neurotropism associated with the wild type VSV glycoprotein.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Neurosurgery, Yale University School of Medicine, 333 Cedar St, New Haven, CT 06520, United States
| | - Guochao Mao
- Department of Neurosurgery, Yale University School of Medicine, 333 Cedar St, New Haven, CT 06520, United States
| | - Anthony N van den Pol
- Department of Neurosurgery, Yale University School of Medicine, 333 Cedar St, New Haven, CT 06520, United States.
| |
Collapse
|
40
|
Suksanpaisan L, Xu R, Tesfay MZ, Bomidi C, Hamm S, Vandergaast R, Jenks N, Steele MB, Ota-Setlik A, Akhtar H, Luckay A, Nowak R, Peng KW, Eldridge JH, Clarke DK, Russell SJ, Diaz RM. Preclinical Development of Oncolytic Immunovirotherapy for Treatment of HPV POS Cancers. MOLECULAR THERAPY-ONCOLYTICS 2018; 10:1-13. [PMID: 29998190 PMCID: PMC6037044 DOI: 10.1016/j.omto.2018.05.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 05/25/2018] [Indexed: 12/18/2022]
Abstract
Immunotherapy for HPVPOS malignancies is attractive because well-defined, viral, non-self tumor antigens exist as targets. Several approaches to vaccinate therapeutically against HPV E6 and E7 antigens have been adopted, including viral platforms such as VSV. A major advantage of VSV expressing these antigens is that VSV also acts as an oncolytic virus, leading to direct tumor cell killing and induction of effective anti-E6 and anti-E7 T cell responses. We have also shown that addition of immune adjuvant genes, such as IFNβ, further enhances safety and/or efficacy of VSV-based oncolytic immunovirotherapies. However, multiple designs of the viral vector are possible—with respect to levels of immunogen expression and method of virus attenuation—and optimal designs have not previously been tested head-to-head. Here, we tested three different VSV engineered to express a non-oncogenic HPV16 E7/6 fusion protein for their immunotherapeutic and oncolytic properties. We assessed their profiles of efficacy and toxicity against HPVPOS and HPVNEG murine tumor models and determined the optimal route of administration. Our data show that VSV is an excellent platform for the oncolytic immunovirotherapy of tumors expressing HPV target antigens, combining a balance of efficacy and safety suitable for evaluation in a first-in-human clinical trial.
Collapse
Affiliation(s)
| | - Rong Xu
- Profectus Biosciences, Inc., Pearl River, NY 10965, USA
| | | | | | - Stefan Hamm
- Profectus Biosciences, Inc., Pearl River, NY 10965, USA
| | | | - Nathan Jenks
- Toxicology and Pharmacology Laboratory, Mayo Clinic, Rochester, MN 55905, USA
| | - Michael B Steele
- Toxicology and Pharmacology Laboratory, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Hinna Akhtar
- Profectus Biosciences, Inc., Pearl River, NY 10965, USA
| | - Amara Luckay
- Profectus Biosciences, Inc., Pearl River, NY 10965, USA
| | - Rebecca Nowak
- Profectus Biosciences, Inc., Pearl River, NY 10965, USA
| | - Kah Whye Peng
- Toxicology and Pharmacology Laboratory, Mayo Clinic, Rochester, MN 55905, USA.,Vyriad, Inc., Rochester, MN 55902, USA.,Deparment of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | - Stephen J Russell
- Vyriad, Inc., Rochester, MN 55902, USA.,Deparment of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | | |
Collapse
|
41
|
Luo L, Yang Y, Du T, Kang T, Xiong M, Cheng H, Liu Y, Wu Y, Li Y, Chen Y, Zhang Q, Liu X, Wei X, Mi P, She Z, Gao G, Wei Y, Gou M. Targeted Nanoparticle-Mediated Gene Therapy Mimics Oncolytic Virus for Effective Melanoma Treatment. ADVANCED FUNCTIONAL MATERIALS 2018. [DOI: 10.1002/adfm.201800173] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Li Luo
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University; Chengdu 610041 China
| | - Yuping Yang
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University; Chengdu 610041 China
| | - Ting Du
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University; Chengdu 610041 China
| | - Tianyi Kang
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University; Chengdu 610041 China
| | - Meimei Xiong
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University; Chengdu 610041 China
| | - Hao Cheng
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University; Chengdu 610041 China
| | - Yu Liu
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University; Chengdu 610041 China
| | - Yujiao Wu
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University; Chengdu 610041 China
| | - Yang Li
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University; Chengdu 610041 China
| | - Yuwen Chen
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University; Chengdu 610041 China
| | - Qianqian Zhang
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University; Chengdu 610041 China
| | - Xuan Liu
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University; Chengdu 610041 China
| | - Xiawei Wei
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University; Chengdu 610041 China
| | - Peng Mi
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University; Chengdu 610041 China
| | - Zhigang She
- Department of Cardiology; Renmin Hospital of Wuhan University and Cardiovascular Research Institute; Wuhan University; Wuhan 430060 China
| | - Guangping Gao
- Horae Gene Therapy Center; University of Massachusetts Medical School; Worcester MA 01605 USA
| | - Yuquan Wei
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University; Chengdu 610041 China
| | - Maling Gou
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University; Chengdu 610041 China
| |
Collapse
|
42
|
Zemp F, Rajwani J, Mahoney DJ. Rhabdoviruses as vaccine platforms for infectious disease and cancer. Biotechnol Genet Eng Rev 2018; 34:122-138. [PMID: 29781359 DOI: 10.1080/02648725.2018.1474320] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The family Rhabdoviridae (RV) comprises a large, genetically diverse collection of single-stranded, negative sense RNA viruses from the order Mononegavirales. Several RV members are being developed as live-attenuated vaccine vectors for the prevention or treatment of infectious disease and cancer. These include the prototype recombinant Vesicular Stomatitis Virus (rVSV) and the more recently developed recombinant Maraba Virus, both species within the genus Vesiculoviridae. A relatively strong safety profile in humans, robust immunogenicity and genetic malleability are key features that make the RV family attractive vaccine platforms. Currently, the rVSV vector is in preclinical development for vaccination against numerous high-priority infectious diseases, with clinical evaluation underway for HIV/AIDS and Ebola virus disease. Indeed, the success of the rVSV-ZEBOV vaccine during the 2014-15 Ebola virus outbreak in West Africa highlights the therapeutic potential of rVSV as a vaccine vector for acute, life-threatening viral illnesses. The rVSV and rMaraba platforms are also being tested as 'oncolytic' cancer vaccines in a series of phase 1-2 clinical trials, after being proven effective at eliciting immune-mediated tumour regression in preclinical mouse models. In this review, we discuss the biological and genetic features that make RVs attractive vaccine platforms and the development and ongoing testing of rVSV and rMaraba strains as vaccine vectors for infectious disease and cancer.
Collapse
Affiliation(s)
- Franz Zemp
- a Alberta Children's Hospital Research Institute , Calgary , Canada.,b Charbonneau Cancer Research Institute , Calgary , Canada
| | - Jahanara Rajwani
- a Alberta Children's Hospital Research Institute , Calgary , Canada.,b Charbonneau Cancer Research Institute , Calgary , Canada.,d Department of Biochemistry and Molecular Biology, Faculty of Medicine , University of Calgary , Calgary , Canada
| | - Douglas J Mahoney
- a Alberta Children's Hospital Research Institute , Calgary , Canada.,b Charbonneau Cancer Research Institute , Calgary , Canada.,c Department of Microbiology, Immunology and Infectious Disease , Faculty of Medicine , University of Calgary , Calgary , Canada.,d Department of Biochemistry and Molecular Biology, Faculty of Medicine , University of Calgary , Calgary , Canada
| |
Collapse
|
43
|
Russell SJ, Barber GN. Oncolytic Viruses as Antigen-Agnostic Cancer Vaccines. Cancer Cell 2018; 33:599-605. [PMID: 29634947 PMCID: PMC5918693 DOI: 10.1016/j.ccell.2018.03.011] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 02/26/2018] [Accepted: 03/09/2018] [Indexed: 02/07/2023]
Abstract
Selective destruction of neoplastic tissues by oncolytic viruses (OVs) leads to antigen-agnostic boosting of neoantigen-specific cytotoxic T lymphocyte (CTL) responses, making OVs ideal companions for checkpoint blockade therapy. Here we discuss the mechanisms whereby OVs modulate both adjuvanticity and antigenicity of tumor cells. Suppression of antitumor immunity after OV therapy has not been observed, possibly because viral antigen expression diminishes as the antiviral response matures, thereby progressively honing the CTL response to tumor neoantigens. By combining direct in situ tumor destruction with the ability to boost antitumor immunity, OVs also have the potential to be powerful standalone cancer therapies.
Collapse
Affiliation(s)
- Stephen J Russell
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA.
| | - Glen N Barber
- Department of Cell Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
44
|
Franz KM, Neidermyer WJ, Tan YJ, Whelan SPJ, Kagan JC. STING-dependent translation inhibition restricts RNA virus replication. Proc Natl Acad Sci U S A 2018; 115:E2058-E2067. [PMID: 29440426 PMCID: PMC5834695 DOI: 10.1073/pnas.1716937115] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In mammalian cells, IFN responses that occur during RNA and DNA virus infections are activated by distinct signaling pathways. The RIG-I-like-receptors (RLRs) bind viral RNA and engage the adaptor MAVS (mitochondrial antiviral signaling) to promote IFN expression, whereas cGAS (cGMP-AMP synthase) binds viral DNA and activates an analogous pathway via the protein STING (stimulator of IFN genes). In this study, we confirm that STING is not necessary to induce IFN expression during RNA virus infection but also find that STING is required to restrict the replication of diverse RNA viruses. The antiviral activities of STING were not linked to its ability to regulate basal expression of IFN-stimulated genes, activate transcription, or autophagy. Using vesicular stomatitis virus as a model, we identified a requirement of STING to inhibit translation during infection and upon transfection of synthetic RLR ligands. This inhibition occurs at the level of translation initiation and restricts the production of viral and host proteins. The inability to restrict translation rendered STING-deficient cells 100 times more likely to support productive viral infections than wild-type counterparts. Genetic analysis linked RNA sensing by RLRs to STING-dependent translation inhibition, independent of MAVS. Thus, STING has dual functions in host defense, regulating protein synthesis to prevent RNA virus infection and regulating IFN expression to restrict DNA viruses.
Collapse
Affiliation(s)
- Kate M Franz
- Division of Gastroenterology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - William J Neidermyer
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| | - Yee-Joo Tan
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore 117545
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore 138673
| | - Sean P J Whelan
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| | - Jonathan C Kagan
- Division of Gastroenterology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115;
| |
Collapse
|
45
|
Oncotargeting by Vesicular Stomatitis Virus (VSV): Advances in Cancer Therapy. Viruses 2018; 10:v10020090. [PMID: 29473868 PMCID: PMC5850397 DOI: 10.3390/v10020090] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 02/12/2018] [Accepted: 02/15/2018] [Indexed: 12/28/2022] Open
Abstract
Modern oncotherapy approaches are based on inducing controlled apoptosis in tumor cells. Although a number of apoptosis-induction approaches are available, site-specific delivery of therapeutic agents still remain the biggest hurdle in achieving the desired cancer treatment benefit. Additionally, systemic treatment-induced toxicity remains a major limiting factor in chemotherapy. To specifically address drug-accessibility and chemotherapy side effects, oncolytic virotherapy (OV) has emerged as a novel cancer treatment alternative. In OV, recombinant viruses with higher replication capacity and stronger lytic properties are being considered for tumor cell-targeting and subsequent cell lysing. Successful application of OVs lies in achieving strict tumor-specific tropism called oncotropism, which is contingent upon the biophysical interactions of tumor cell surface receptors with viral receptors and subsequent replication of oncolytic viruses in cancer cells. In this direction, few viral vector platforms have been developed and some of these have entered pre-clinical/clinical trials. Among these, the Vesicular stomatitis virus (VSV)-based platform shows high promise, as it is not pathogenic to humans. Further, modern molecular biology techniques such as reverse genetics tools have favorably advanced this field by creating efficient recombinant VSVs for OV; some have entered into clinical trials. In this review, we discuss the current status of VSV based oncotherapy, challenges, and future perspectives regarding its therapeutic applications in the cancer treatment.
Collapse
|
46
|
Sahu SK, Kumar M. Application of Oncolytic Virus as a Therapy of Cancer. Microb Biotechnol 2018. [DOI: 10.1007/978-981-10-7140-9_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
47
|
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: 65] [Impact Index Per Article: 9.3] [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
|
48
|
Fountzilas C, Patel S, Mahalingam D. Review: Oncolytic virotherapy, updates and future directions. Oncotarget 2017; 8:102617-102639. [PMID: 29254276 PMCID: PMC5731986 DOI: 10.18632/oncotarget.18309] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 05/03/2017] [Indexed: 12/14/2022] Open
Abstract
Oncolytic viruses (OVs) are viral strains that can infect and kill malignant cells while spare their normal counterparts. OVs can access cells through binding to receptors on their surface or through fusion with the plasma membrane and establish a lytic cycle in tumors, while leaving normal tissue essentially unharmed. Multiple viruses have been investigated in humans for the past century. IMLYGIC™ (T-VEC/Talimogene Laherparepvec), a genetically engineered Herpes Simplex Virus, is the first OV approved for use in the United States and the European Union for patients with locally advanced or non-resectable melanoma. Although OVs have a favorable toxicity profile and are impressively active anticancer agents in vitro and in vivo the majority of OVs have limited clinical efficacy as a single agent. While a virus-induced antitumor immune response can enhance oncolysis, when OVs are used systemically, the antiviral immune response can prevent the virus reaching the tumor tissue and having a therapeutic effect. Intratumoral administration can provide direct access to tumor tissue and be beneficial in reducing side effects. Immune checkpoint stimulation in tumor tissue has been noted after OV therapy and can be a natural response to viral-induced oncolysis. Also for immune checkpoint inhibition to be effective in treating cancer, an immune response to tumor neoantigens and an inflamed tumor microenvironment are required, both of which treatment with an OV may provide. Therefore, direct and indirect mechanisms of tumor killing provide rationale for clinical trials investigating the combination of OVs other forms of cancer therapy, including immune checkpoint inhibition.
Collapse
Affiliation(s)
- Christos Fountzilas
- The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Sukeshi Patel
- The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | | |
Collapse
|
49
|
Naik S, Galyon GD, Jenks NJ, Steele MB, Miller AC, Allstadt SD, Suksanpaisan L, Peng KW, Federspiel MJ, Russell SJ, LeBlanc AK. Comparative Oncology Evaluation of Intravenous Recombinant Oncolytic Vesicular Stomatitis Virus Therapy in Spontaneous Canine Cancer. Mol Cancer Ther 2017; 17:316-326. [PMID: 29158470 DOI: 10.1158/1535-7163.mct-17-0432] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 07/13/2017] [Accepted: 11/03/2017] [Indexed: 12/22/2022]
Abstract
Clinical translation of intravenous therapies to treat disseminated or metastatic cancer is imperative. Comparative oncology, the evaluation of novel cancer therapies in animals with spontaneous cancer, can be utilized to inform and accelerate clinical translation. Preclinical murine studies demonstrate that single-shot systemic therapy with a vesicular stomatitis virus (VSV)-IFNβ-NIS, a novel recombinant oncolytic VSV, can induce curative remission in tumor-bearing mice. Clinical translation of VSV-IFNβ-NIS therapy is dependent on comprehensive assessment of clinical toxicities, virus shedding, pharmacokinetics, and efficacy in clinically relevant models. Dogs spontaneously develop cancer with comparable etiology, clinical progression, and response to therapy as human malignancies. A comparative oncology study was carried out to investigate feasibility and tolerability of intravenous oncolytic VSV-IFNβ-NIS therapy in pet dogs with spontaneous cancer. Nine dogs with various malignancies were treated with a single intravenous dose of VSV-IFNβ-NIS. Two dogs with high-grade peripheral T-cell lymphoma had rapid but transient remission of disseminated disease and transient hepatotoxicity that resolved spontaneously. There was no shedding of infectious virus. Correlative pharmacokinetic studies revealed elevated levels of VSV RNA in blood in dogs with measurable disease remission. This is the first evaluation of intravenous oncolytic virus therapy for spontaneous canine cancer, demonstrating that VSV-IFNβ-NIS is well-tolerated and safe in dogs with advanced or metastatic disease. This approach has informed clinical translation, including dose and target indication selection, leading to a clinical investigation of intravenous VSV-IFNβ-NIS therapy, and provided preliminary evidence of clinical efficacy and potential biomarkers that correlate with therapeutic response. Mol Cancer Ther; 17(1); 316-26. ©2017 AACR.
Collapse
Affiliation(s)
- Shruthi Naik
- Department of Molecular Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota.,Vyriad, Inc., Rochester, Minnesota
| | - Gina D Galyon
- Small Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee
| | - Nathan J Jenks
- Toxicology and Pharmacology Laboratory, Mayo Clinic, Rochester, Minnesota
| | - Michael B Steele
- Toxicology and Pharmacology Laboratory, Mayo Clinic, Rochester, Minnesota
| | - Amber C Miller
- Department of Molecular Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Sara D Allstadt
- Small Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee
| | | | - Kah Whye Peng
- Toxicology and Pharmacology Laboratory, Mayo Clinic, Rochester, Minnesota
| | - Mark J Federspiel
- Viral Vector Production Laboratory, Mayo Clinic, Rochester, Minnesota
| | - Stephen J Russell
- Department of Molecular Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota.,Vyriad, Inc., Rochester, Minnesota
| | - Amy K LeBlanc
- Small Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee.
| |
Collapse
|
50
|
Velazquez-Salinas L, Naik S, Pauszek SJ, Peng KW, Russell SJ, Rodriguez LL. Oncolytic Recombinant Vesicular Stomatitis Virus (VSV) Is Nonpathogenic and Nontransmissible in Pigs, a Natural Host of VSV. HUM GENE THER CL DEV 2017; 28:108-115. [PMID: 28514874 DOI: 10.1089/humc.2017.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Vesicular stomatitis virus (VSV) is a negative-stranded RNA virus that naturally causes disease in livestock including horses, cattle and pigs. The two main identified VSV serotypes are New Jersey (VSNJV) and Indiana (VSIV). VSV is a rapidly replicating, potently immunogenic virus that has been engineered to develop novel oncolytic therapies for cancer treatment. Swine are a natural host for VSV and provide a relevant and well-established model, amenable to biological sampling to monitor virus shedding and neutralizing antibodies. Previous reports have documented the pathogenicity and transmissibility of wild-type isolates and recombinant strains of VSIV and VSNJV using the swine model. Oncolytic VSV engineered to express interferon-beta (IFNβ) and the sodium iodide symporter (NIS), VSV-IFNβ-NIS, has been shown to be a potent new therapeutic agent inducing rapid and durable tumor remission following systemic therapy in preclinical mouse models. VSV-IFNβ-NIS is currently undergoing clinical evaluation for the treatment of advanced cancer in human and canine patients. To support clinical studies and comprehensively assess the risk of transmission to susceptible species, we tested the pathogenicity and transmissibility of oncolytic VSV-IFNβ-NIS using the swine model. Following previously established protocols to evaluate VSV pathogenicity, intradermal inoculation with 107 TCID50 VSV-IFNβ-NIS caused no observable symptoms in pigs. There was no detectable shedding of infectious virus in VSV-IFNβ-NIS in biological excreta of inoculated pigs or exposed naive pigs kept in direct contact throughout the experiment. VSV-IFNβ-NIS inoculated pigs became seropositive for VSV antibodies, while contact pigs displayed no symptoms of VSV infection, and importantly did not seroconvert. These data indicate that oncolytic VSV is both nonpathogenic and not transmissible in pigs, a natural host. These findings support further clinical development of oncolytic VSV-IFNβ-NIS as a safe therapeutic for human and canine cancer.
Collapse
Affiliation(s)
- Lauro Velazquez-Salinas
- 1 United States Department of Agriculture, Agricultural Research Services , Foreign Animal Disease Research Unit, Plum Island, New York
| | - Shruthi Naik
- 2 Vyriad, Inc., Rochester Minnesota.,3 Department of Molecular Medicine, Mayo Clinic College of Medicine , Rochester, Minnesota
| | - Steven J Pauszek
- 1 United States Department of Agriculture, Agricultural Research Services , Foreign Animal Disease Research Unit, Plum Island, New York
| | - Kah-Whye Peng
- 3 Department of Molecular Medicine, Mayo Clinic College of Medicine , Rochester, Minnesota.,4 Toxicology and Pharmacology Laboratory, Mayo Clinic College of Medicine , Rochester, Minnesota
| | | | - Luis L Rodriguez
- 1 United States Department of Agriculture, Agricultural Research Services , Foreign Animal Disease Research Unit, Plum Island, New York
| |
Collapse
|