1
|
Rafiq Z, Kang M, Barsoumian HB, Manzar GS, Hu Y, Leuschner C, Huang A, Masrorpour F, Lu W, Puebla-Osorio N, Welsh JW. Enhancing immunotherapy efficacy with synergistic low-dose radiation in metastatic melanoma: current insights and prospects. J Exp Clin Cancer Res 2025; 44:31. [PMID: 39881333 PMCID: PMC11781074 DOI: 10.1186/s13046-025-03281-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 01/09/2025] [Indexed: 01/31/2025] Open
Abstract
Recent advances in oncology research have highlighted the promising synergy between low-dose radiation therapy (LDRT) and immunotherapies, with growing evidence highlighting the unique benefits of the combination. LDRT has emerged as a potent tool for stimulating the immune system, triggering systemic antitumor effects by remodeling the tumor microenvironment. Notably, LDRT demonstrates remarkable efficacy even in challenging metastatic sites such as the liver (uveal) and brain (cutaneous), particularly in advanced melanoma stages. The increasing interest in utilizing LDRT for secondary metastatic sites of uveal, mucosal, or cutaneous melanomas underscores its potential efficacy in combination with various immunotherapies. This comprehensive review traverses the journey from laboratory research to clinical applications, elucidating LDRT's immunomodulatory role on the tumor immune microenvironment (TIME) and systemic immune responses. We meticulously examine the preclinical evidence and ongoing clinical trials, throwing light on the promising prospects of LDRT as a complementary therapy in melanoma treatment. Furthermore, we explore the challenges associated with LDRT's integration into combination therapies, addressing crucial factors such as optimal dosage, fractionation, treatment frequency, and synergy with other pharmacological agents. Considering its low toxicity profile, LDRT presents a compelling case for application across multiple lesions, augmenting the antitumor immune response in poly-metastatic disease scenarios. The convergence of LDRT with other disciplines holds immense potential for developing novel radiotherapy-combined modalities, paving the way for more effective and personalized treatment strategies in melanoma and beyond. Moreover, the dose-related toxicities of immunotherapies may be reduced by synergistic amplification of antitumor efficacy with LDRT.
Collapse
Affiliation(s)
- Zahid Rafiq
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 500 W. University Ave, El Paso, TX, 79968, USA
| | - Mingyo Kang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Hampartsoum B Barsoumian
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Gohar S Manzar
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yun Hu
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Carola Leuschner
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ailing Huang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Fatemeh Masrorpour
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Weiqin Lu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 500 W. University Ave, El Paso, TX, 79968, USA
| | - Nahum Puebla-Osorio
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - James W Welsh
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
2
|
Rajwani J, Vishnevskiy D, Turk M, Naumenko V, Gafuik C, Kim DS, Mah LK, Snelling S, Gonzales GA, Xue J, Chanda A, Potts KG, Todesco HM, Lau KCK, Hildebrand KM, Chan JA, Liao S, Monument MJ, Hyrcza M, Bose P, Jenne CN, Canton J, Zemp FJ, Mahoney DJ. VSV ∆M51 drives CD8 + T cell-mediated tumour regression through infection of both cancer and non-cancer cells. Nat Commun 2024; 15:9933. [PMID: 39548070 PMCID: PMC11567966 DOI: 10.1038/s41467-024-54111-6] [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: 10/11/2023] [Accepted: 11/02/2024] [Indexed: 11/17/2024] Open
Abstract
Oncolytic viruses (OV) are designed to selectively infect and kill cancer cells, while simultaneously eliciting antitumour immunity. The mechanism is expected to originate from infected cancer cells. However, recent reports of tumour regression unaccompanied by cancer cell infection suggest a more complex mechanism of action. Here, we engineered vesicular stomatitis virus (VSV)ΔM51-sensitive and VSVΔM51-resistant tumour lines to elucidate the role of OV-infected cancer and non-cancer cells. We found that, while cancer cell infections elicit oncolysis and antitumour immunity as expected, infection of non-cancer cells alone can also contribute to tumour regression. This effect is partly attributed to the systemic production of cytokines that promote dendritic cell (DC) activation, migration and antigen cross-presentation, leading to magnified antitumour CD8+ T cell activation and tumour regression. Such OV-induced antitumour immunity is complementary to PD-1 blockade. Overall, our results reveal mechanistic insights into OV-induced antitumour immunity that can be leveraged to improve OV-based therapeutics.
Collapse
Affiliation(s)
- Jahanara Rajwani
- Arnie Charbonneau Cancer Institute; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Alberta Children's Hospital Research Institute; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine; University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Daniil Vishnevskiy
- Arnie Charbonneau Cancer Institute; University of Calgary, Calgary, AB, T2N 4N1, Canada
- The Calvin, Joan and Phoebe Snyder Institute for Chronic Disease; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine; University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Madison Turk
- Arnie Charbonneau Cancer Institute; University of Calgary, Calgary, AB, T2N 4N1, Canada
- The Calvin, Joan and Phoebe Snyder Institute for Chronic Disease; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine; University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Victor Naumenko
- Arnie Charbonneau Cancer Institute; University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Chris Gafuik
- Arnie Charbonneau Cancer Institute; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Alberta Children's Hospital Research Institute; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine; University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Dae-Sun Kim
- Arnie Charbonneau Cancer Institute; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Alberta Children's Hospital Research Institute; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine; University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Laura K Mah
- Arnie Charbonneau Cancer Institute; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Alberta Children's Hospital Research Institute; University of Calgary, Calgary, AB, T2N 4N1, Canada
- The Calvin, Joan and Phoebe Snyder Institute for Chronic Disease; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine; University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Shannon Snelling
- Arnie Charbonneau Cancer Institute; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Alberta Children's Hospital Research Institute; University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Gerone A Gonzales
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Faculty of Veterinary Medicine; University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Jingna Xue
- The Calvin, Joan and Phoebe Snyder Institute for Chronic Disease; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine; University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Ayan Chanda
- Arnie Charbonneau Cancer Institute; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine; University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Kyle G Potts
- Arnie Charbonneau Cancer Institute; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Alberta Children's Hospital Research Institute; University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Hayley M Todesco
- Arnie Charbonneau Cancer Institute; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Alberta Children's Hospital Research Institute; University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Keith C K Lau
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine; University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Karys M Hildebrand
- Arnie Charbonneau Cancer Institute; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Surgery, Cumming School of Medicine; University of Calgary, Calgary, AB, T2N 4N1, Canada
- McCaig Bone and Joint Institute, Cumming School of Medicine; University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Jennifer A Chan
- Arnie Charbonneau Cancer Institute; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Pathology and Laboratory Medicine; University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Shan Liao
- The Calvin, Joan and Phoebe Snyder Institute for Chronic Disease; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine; University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Michael J Monument
- Arnie Charbonneau Cancer Institute; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Surgery, Cumming School of Medicine; University of Calgary, Calgary, AB, T2N 4N1, Canada
- McCaig Bone and Joint Institute, Cumming School of Medicine; University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Martin Hyrcza
- Arnie Charbonneau Cancer Institute; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Pathology and Laboratory Medicine; University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Pinaki Bose
- Arnie Charbonneau Cancer Institute; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Oncology; University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Craig N Jenne
- The Calvin, Joan and Phoebe Snyder Institute for Chronic Disease; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine; University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Johnathan Canton
- The Calvin, Joan and Phoebe Snyder Institute for Chronic Disease; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Faculty of Veterinary Medicine; University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Franz J Zemp
- Arnie Charbonneau Cancer Institute; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Alberta Children's Hospital Research Institute; University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine; University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Douglas J Mahoney
- Arnie Charbonneau Cancer Institute; University of Calgary, Calgary, AB, T2N 4N1, Canada.
- Alberta Children's Hospital Research Institute; University of Calgary, Calgary, AB, T2N 4N1, Canada.
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine; University of Calgary, Calgary, AB, T2N 4N1, Canada.
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine; University of Calgary, Calgary, AB, T2N 4N1, Canada.
| |
Collapse
|
3
|
Raimondi V, Vescovini R, Dessena M, Donofrio G, Storti P, Giuliani N. Oncolytic viruses: a potential breakthrough immunotherapy for multiple myeloma patients. Front Immunol 2024; 15:1483806. [PMID: 39539548 PMCID: PMC11557349 DOI: 10.3389/fimmu.2024.1483806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Oncolytic virotherapy represents an innovative and promising approach for the treatment of cancer, including multiple myeloma (MM), a currently incurable plasma cell (PC) neoplasm. Despite the advances that new therapies, particularly immunotherapy, have been made, relapses still occur in MM patients, highlighting the medical need for new treatment options. Oncolytic viruses (OVs) preferentially infect and destroy cancer cells, exerting a direct and/or indirect cytopathic effect, combined with a modulation of the tumor microenvironment leading to an activation of the immune system. Both naturally occurring and genetically modified viruses have demonstrated significant preclinical effects against MM cells. Currently, the OVs genetically modified measles virus strains, reovirus, and vesicular stomatitis virus are employed in clinical trials for MM. Nevertheless, significant challenges remain, including the efficiency of the virus delivery to the tumor, overcoming antiviral immune responses, and the specificity of the virus for MM cells. Different strategies are being explored to optimize OV therapy, including combining it with standard treatments and targeted therapies to enhance efficacy. This review will provide a comprehensive analysis of the mechanism of action of the different OVs, and preclinical and clinical evidence, focusing on the role of oncolytic virotherapy as a new possible immunotherapeutic approach also in combination with the current therapeutic armamentarium and underlying the future directions in the context of MM treatments.
Collapse
Affiliation(s)
- Vincenzo Raimondi
- Laboratory of Hematology, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Rosanna Vescovini
- Laboratory of Hematology, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Mattia Dessena
- Laboratory of Hematology, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Gaetano Donofrio
- Department of Medical-Veterinary Science, University of Parma, Parma, Italy
| | - Paola Storti
- Laboratory of Hematology, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Nicola Giuliani
- Laboratory of Hematology, Department of Medicine and Surgery, University of Parma, Parma, Italy
- Multiple Myeloma and Monoclonal Gammopathy Program, Department of Onco-Hematology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
- Hematology Unit, Department of Onco-Hematology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| |
Collapse
|
4
|
Robinson SI, Rochell RE, Penza V, Naik S. Translation of oncolytic viruses in sarcoma. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200822. [PMID: 39040851 PMCID: PMC11261849 DOI: 10.1016/j.omton.2024.200822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Sarcomas are a rare and highly diverse group of malignancies of mesenchymal origin. While sarcomas are generally considered resistant to immunotherapy, recent studies indicate subtype-specific differences in clinical response to checkpoint inhibitors (CPIs) that are associated with distinct immune phenotypes present in sarcoma subtypes. Oncolytic viruses (OVs) are designed to selectively infect and kill tumor cells and induce intratumoral immune infiltration, enhancing immunogenicity and thereby sensitizing tumors to immunotherapy. Herein we review the accumulated clinical data evaluating OVs in sarcoma. Small numbers of patients with sarcoma were enrolled in early-stage OV trials as part of larger solid tumor cohorts demonstrating safety but providing limited insight into the biological effects due to the low patient numbers and lack of histologic grouping. Several recent studies have investigated talimogene laherparepvec (T-VEC), an approved oncolytic herpes simplex virus (HSV-1), in combination therapy regimens in sarcoma patient cohorts. These studies have shown promising responses in heavily pre-treated and immunotherapy-resistant patients associated with increased intratumoral immune infiltration. As new and more potent OVs enter the clinical arena, prospective evaluation in subtype-specific cohorts with correlative studies to define biomarkers of response will be critical to advancing this promising approach for sarcoma therapy.
Collapse
Affiliation(s)
- Steven I. Robinson
- Division of Medical Oncology, Mayo Clinic, Rochester, MN 55902, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Roya E. Rochell
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Velia Penza
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Shruthi Naik
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
5
|
Gao X, Liu J, Sun R, Zhang J, Cao X, Zhang Y, Zhao M. Alliance between titans: combination strategies of CAR-T cell therapy and oncolytic virus for the treatment of hematological malignancies. Ann Hematol 2024; 103:2569-2589. [PMID: 37853078 DOI: 10.1007/s00277-023-05488-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 09/28/2023] [Indexed: 10/20/2023]
Abstract
There have been several clinical studies using chimeric antigen receptor (CAR)-T cell therapy for different hematological malignancies. It has transformed the therapy landscape for hematologic malignancies dramatically. Nonetheless, in acute myeloid leukemia (AML) and T cell malignancies, it still has a dismal prognosis. Even in the most promising locations, recurrence with CAR-T treatment remains a big concern. Oncolytic viruses (OVs) can directly lyse tumor cells or cause immune responses, and they can be manipulated to create therapeutic proteins, increasing anticancer efficacy. Oncolytic viruses have been proven in a rising number of studies to be beneficial in hematological malignancies. There are limitations that cannot be avoided by using either treatment alone, and the combination of CAR-T cell therapy and oncolytic virus therapy may complement the disadvantages of individual application, enhance the advantages of their respective treatment methods and improve the treatment effect. The alternatives for combining two therapies in hematological malignancies are discussed in this article.
Collapse
Affiliation(s)
- Xuejin Gao
- Emergency, Tianjin First Central Hospital, Tianjin, 300192, China
| | - Jile Liu
- First Center Clinic College of Tianjin Medical University, Tianjin, 300192, China
| | - Rui Sun
- Nankai University School of Medicine, Tianjin, 300192, China
| | - Jingkun Zhang
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Xinping Cao
- First Center Clinic College of Tianjin Medical University, Tianjin, 300192, China
| | - Yi Zhang
- First Center Clinic College of Tianjin Medical University, Tianjin, 300192, China
| | - Mingfeng Zhao
- Department of Hematology, Tianjin First Central Hospital, Tianjin, 300192, China.
| |
Collapse
|
6
|
Lin C, Teng W, Tian Y, Li S, Xia N, Huang C. Immune landscape and response to oncolytic virus-based immunotherapy. Front Med 2024; 18:411-429. [PMID: 38453818 DOI: 10.1007/s11684-023-1048-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 11/15/2023] [Indexed: 03/09/2024]
Abstract
Oncolytic virus (OV)-based immunotherapy has emerged as a promising strategy for cancer treatment, offering a unique potential to selectively target malignant cells while sparing normal tissues. However, the immunosuppressive nature of tumor microenvironment (TME) poses a substantial hurdle to the development of OVs as effective immunotherapeutic agents, as it restricts the activation and recruitment of immune cells. This review elucidates the potential of OV-based immunotherapy in modulating the immune landscape within the TME to overcome immune resistance and enhance antitumor immune responses. We examine the role of OVs in targeting specific immune cell populations, including dendritic cells, T cells, natural killer cells, and macrophages, and their ability to alter the TME by inhibiting angiogenesis and reducing tumor fibrosis. Additionally, we explore strategies to optimize OV-based drug delivery and improve the efficiency of OV-mediated immunotherapy. In conclusion, this review offers a concise and comprehensive synopsis of the current status and future prospects of OV-based immunotherapy, underscoring its remarkable potential as an effective immunotherapeutic agent for cancer treatment.
Collapse
Affiliation(s)
- Chaolong Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, 361102, China
| | - Wenzhong Teng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, 361102, China
| | - Yang Tian
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, 361102, China
| | - Shaopeng Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, 361102, China
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China.
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, 361102, China.
| | - Chenghao Huang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China.
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, 361102, China.
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
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. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.16.533664. [PMID: 37131624 PMCID: PMC10153185 DOI: 10.1101/2023.04.16.533664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
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 described 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 RNAseq 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
|
10
|
Dingli D. Radiovirotherapy at twenty. Mol Ther Oncolytics 2023; 29:127-128. [PMID: 37260766 PMCID: PMC10227365 DOI: 10.1016/j.omto.2023.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023] Open
Affiliation(s)
- David Dingli
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
11
|
Watters CR, Barro O, Elliott NM, Zhou Y, Gabere M, Raupach E, Baker AT, Barrett MT, Buetow KH, Jacobs B, Seetharam M, Borad MJ, Nagalo BM. Multi-modal efficacy of a chimeric vesiculovirus expressing the Morreton glycoprotein in sarcoma. Mol Ther Oncolytics 2023; 29:4-14. [PMID: 36969560 PMCID: PMC10033453 DOI: 10.1016/j.omto.2023.02.009] [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: 09/29/2022] [Accepted: 02/24/2023] [Indexed: 03/06/2023] Open
Abstract
Vesiculoviruses are attractive oncolytic virus platforms due to their rapid replication, appreciable transgene capacity, broad tropism, limited preexisting immunity, and tumor selectivity through type I interferon response defects in malignant cells. We developed a synthetic chimeric virus (VMG) expressing the glycoprotein (G) from Morreton virus (MorV) and utilizing the remaining structural genes from vesicular stomatitis virus (VSV). VMG exhibited in vitro efficacy by inducing oncolysis in a broad range of sarcoma subtypes across multiple species. Notably, all cell lines tested showed the ability of VMG to yield productive infection with rapid replication kinetics and induction of apoptosis. Furthermore, pilot safety evaluations of VMG in immunocompetent, non-tumor-bearing mice showed an absence of toxicity with intranasal doses as high as 1e10 50% tissue culture infectious dose (TCID50)/kg. Locoregional administration of VMG in vivo resulted in tumor reduction in an immunodeficient Ewing sarcoma xenograft at doses as low as 2e5 TCID50. In a murine syngeneic fibrosarcoma model, while no tumor inhibition was achieved with VMG, there was a robust induction of CD8+ T cells within the tumor. The studies described herein establish the promising potential for VMG to be used as a novel oncolytic virotherapy platform with anticancer effects in sarcoma.
Collapse
Affiliation(s)
- Chelsae R. Watters
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Oumar Barro
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Natalie M. Elliott
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Yumei Zhou
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, AZ 85259, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Musa Gabere
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, AZ 85259, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Elizabeth Raupach
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, AZ 85259, USA
| | | | - Michael T. Barrett
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, AZ 85259, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, AZ 85054, USA
| | - Kenneth H. Buetow
- Computational Sciences and Informatics Program for Complex Adaptive System Arizona State University, Tempe, AZ 85281, USA
| | - Bertram Jacobs
- Center for Infectious Diseases and Vaccinology, the Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
| | - Mahesh Seetharam
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, AZ 85259, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, AZ 85054, USA
| | - Mitesh J. Borad
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, AZ 85259, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, AZ 85054, USA
| | - Bolni Marius Nagalo
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- The Winthrop P. Rockefeller Cancer Institute, UAMS, Little Rock, AR 72205, USA
| |
Collapse
|
12
|
Zolaly MA, Mahallawi W, Khawaji ZY, Alahmadi MA. The Clinical Advances of Oncolytic Viruses in Cancer Immunotherapy. Cureus 2023; 15:e40742. [PMID: 37485097 PMCID: PMC10361339 DOI: 10.7759/cureus.40742] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2023] [Indexed: 07/25/2023] Open
Abstract
A promising future for oncology treatment has been brought about by the emergence of a novel approach utilizing oncolytic viruses in cancer immunotherapy. Oncolytic viruses are viruses that have been exploited genetically to assault malignant cells and activate a robust immune response. Several techniques have been developed to endow viruses with an oncolytic activity through genetic engineering. For instance, redirection capsid modification, stimulation of anti-neoplastic immune response, and genetically arming viruses with cytokines such as IL-12. Oncolytic viral clinical outcomes are sought after, particularly in more advanced cancers. The effectiveness and safety profile of the oncolytic virus in clinical studies with or without the combination of standard treatment (chemotherapy, radiotherapy, or primary excision) has been assessed using response evaluation criteria in solid tumors (RECIST). This review will comprehensively outline the most recent clinical applications and provide the results from various phases of clinical trials in a variety of cancers in the latest published literature.
Collapse
Affiliation(s)
- Mohammed A Zolaly
- Pediatric Hematology Oncology, Taibah University, Al-Madinah al-Munawwarah, SAU
| | - Waleed Mahallawi
- Medical Laboratory Technology Department, College of Applied Medical Sciences, Taibah University, Al-Madinah al-Munawwarah, SAU
| | - Zakaria Y Khawaji
- Medicine and Surgery, Taibah University, Al-Madinah al-Munawwarah, SAU
| | | |
Collapse
|
13
|
Thoidingjam S, Sriramulu S, Freytag S, Brown SL, Kim JH, Chetty IJ, Siddiqui F, Movsas B, Nyati S. Oncolytic virus-based suicide gene therapy for cancer treatment: a perspective of the clinical trials conducted at Henry Ford Health. TRANSLATIONAL MEDICINE COMMUNICATIONS 2023; 8:11. [PMID: 37065938 PMCID: PMC10088621 DOI: 10.1186/s41231-023-00144-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 04/04/2023] [Indexed: 06/19/2023]
Abstract
Gene therapy manipulates or modifies a gene that provides a new cellular function to treat or correct a pathological condition, such as cancer. The approach of using gene manipulation to modify patient's cells to improve cancer therapy and potentially find a cure is gaining popularity. Currently, there are 12 gene therapy products approved by US-FDA, EMA and CFDA for cancer management, these include Rexin-G, Gendicine, Oncorine, Provange among other. The Radiation Biology Research group at Henry Ford Health has been actively developing gene therapy approaches for improving clinical outcome in cancer patients. The team was the first to test a replication-competent oncolytic virus armed with a therapeutic gene in humans, to combine this approach with radiation in humans, and to image replication-competent adenoviral gene expression/activity in humans. The adenoviral gene therapy products developed at Henry Ford Health have been evaluated in more than 6 preclinical studies and evaluated in 9 investigator initiated clinical trials treating more than100 patients. Two phase I clinical trials are currently following patients long term and a phase I trial for recurrent glioma was initiated in November 2022. This systematic review provides an overview of gene therapy approaches and products employed for treating cancer patients including the products developed at Henry Ford Health.
Collapse
Affiliation(s)
- Shivani Thoidingjam
- Department of Radiation Oncology, Henry Ford Health, 1 Ford Place, 5D-42, Detroit, MI 48202 USA
| | - Sushmitha Sriramulu
- Department of Radiation Oncology, Henry Ford Health, 1 Ford Place, 5D-42, Detroit, MI 48202 USA
| | - Svend Freytag
- Department of Radiation Oncology, Henry Ford Health, 1 Ford Place, 5D-42, Detroit, MI 48202 USA
| | - Stephen L. Brown
- Department of Radiation Oncology, Henry Ford Health, 1 Ford Place, 5D-42, Detroit, MI 48202 USA
- College of Human Medicine, Michigan State University, East Lansing, MI 48824 USA
| | - Jae Ho Kim
- Department of Radiation Oncology, Henry Ford Health, 1 Ford Place, 5D-42, Detroit, MI 48202 USA
| | - Indrin J. Chetty
- Department of Radiation Oncology, Henry Ford Health, 1 Ford Place, 5D-42, Detroit, MI 48202 USA
| | - Farzan Siddiqui
- Department of Radiation Oncology, Henry Ford Health, 1 Ford Place, 5D-42, Detroit, MI 48202 USA
| | - Benjamin Movsas
- Department of Radiation Oncology, Henry Ford Health, 1 Ford Place, 5D-42, Detroit, MI 48202 USA
- College of Human Medicine, Michigan State University, East Lansing, MI 48824 USA
| | - Shyam Nyati
- Department of Radiation Oncology, Henry Ford Health, 1 Ford Place, 5D-42, Detroit, MI 48202 USA
- College of Human Medicine, Michigan State University, East Lansing, MI 48824 USA
| |
Collapse
|
14
|
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
|
15
|
Naik S, Russell L. The 13 th International Oncolytic Virus Conference: Powerful payloads gain clinical momentum. Mol Ther 2022; 30:1361-1363. [PMID: 35349786 PMCID: PMC9077475 DOI: 10.1016/j.ymthe.2022.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 03/15/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Shruthi Naik
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA; Vyriad, Inc., Rochester, MN, USA.
| | | |
Collapse
|