1
|
Jin H, Zhao YR, Huang F, Hong Z, Jia XY, Wang H, Wang YG. Vaccinia virus-mediated oncolytic immunotherapy: Emerging strategies for gastrointestinal cancer treatment at dawn. Virology 2025; 602:110303. [PMID: 39577274 DOI: 10.1016/j.virol.2024.110303] [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: 08/17/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/24/2024]
Abstract
Oncolytic vaccinia virus (VVs) based immunotherapy is a rapidly developing treatment for gastrointestinal (GI) cancers. Conventional treatments, such as chemotherapy, radiotherapy and surgery achieve good effects in early-stage GI cancers, but effects are limited in advanced disease. Immunotherapy has limited efficacy in GI cancers due to tumor heterogeneity and complex immunosuppressive mechanisms. Oncolytic VV immunotherapy is a novel treatment approach showing promising results in preclinical and clinical trials. Oncolytic VV's intracytoplasmic replication and assembly mechanism, diverse mature forms, and use methods make it extremely safe and versatile for drug delivery. Combining oncolytic VV with conventional therapies and immunotherapy (e.g., ICIs, CAR-T) enhances tumor regression and survival compared to monotherapies. Researchers are establishing response protocols and improvement strategies, rapidly developing VV tumor oncolytic immunotherapy. This article focuses on oncolytic vaccinia development and outlook in gastrointestinal cancer therapy, advantages when combined with other drugs to improve clinical survival, safety, and risk reduction for patients.
Collapse
Affiliation(s)
- Hao Jin
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, Zhejiang Province, China; Oncology Department, Zhejiang Sci-Tech University Shaoxing Academy of Biomedicine, Shaoxing, China
| | - Ya-Ru Zhao
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, Zhejiang Province, China; Oncology Department, Zhejiang Sci-Tech University Shaoxing Academy of Biomedicine, Shaoxing, China
| | - Fang Huang
- Department of Pathology, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China
| | - Zhang Hong
- Department of Respiratory and Critical Care Medicine, Second Medical Center, Chinese PLA General Hospital, Beijing, 100089, China
| | - Xiao-Yuan Jia
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, Zhejiang Province, China
| | - Hui Wang
- Oncology Department, Zhejiang Xiaoshan Hospital, 311201, Hangzhou, China.
| | - Yi-Gang Wang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, Zhejiang Province, China; Oncology Department, Zhejiang Sci-Tech University Shaoxing Academy of Biomedicine, Shaoxing, China.
| |
Collapse
|
2
|
Rojas JJ, Van Hoecke L, Conesa M, Bueno-Merino C, Del Canizo A, Riederer S, Barcia M, Brosinski K, Lehmann MH, Volz A, Saelens X, Sutter G. A new MVA ancestor-derived oncolytic vaccinia virus induces immunogenic tumor cell death and robust antitumor immune responses. Mol Ther 2024; 32:2406-2422. [PMID: 38734899 PMCID: PMC11286824 DOI: 10.1016/j.ymthe.2024.05.014] [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: 10/10/2023] [Revised: 04/10/2024] [Accepted: 05/09/2024] [Indexed: 05/13/2024] Open
Abstract
Vaccinia viruses (VACVs) are versatile therapeutic agents and different features of various VACV strains allow for a broad range of therapeutic applications. Modified VACV Ankara (MVA) is a particularly altered VACV strain that is highly immunogenic, incapable of replicating in mammalian hosts, and broadly used as a safe vector for vaccination. Alternatively, Western Reserve (WR) or Copenhagen (Cop) are VACV strains that efficiently replicate in cancer cells and, therefore, are used to develop oncolytic viruses. However, the immune evasion capacity of WR or Cop hinders their ability to elicit antitumor immune responses, which is crucial for efficacy in the clinic. Here, we describe a new VACV strain named Immune-Oncolytic VACV Ankara (IOVA), which combines efficient replication in cancer cells with induction of immunogenic tumor cell death (ICD). IOVA was engineered from an MVA ancestor and shows superior cytotoxicity in tumor cells. In addition, the IOVA genome incorporates mutations that lead to massive fusogenesis of tumor cells, which contributes to improved antitumor effects. In syngeneic mouse tumor models, the induction of ICD results in robust antitumor immunity directed against tumor neo-epitopes and eradication of large established tumors. These data present IOVA as an improved immunotherapeutic oncolytic vector.
Collapse
Affiliation(s)
- Juan J Rojas
- Immunology Unit, Department of Pathology and Experimental Therapies, School of Medicine, University of Barcelona - UB, 08907 L'Hospitalet de Llobregat, Spain; Immunity, Inflammation, and Cancer Group, Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge - IDIBELL, 08908 L'Hospitalet de Llobregat, Spain; Division of Virology, Institute for Infection Medicine and Zoonoses, Department of Veterinary Sciences, LMU Munich, 85764 Oberschleiβheim, Germany.
| | - Lien Van Hoecke
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Miquel Conesa
- Immunology Unit, Department of Pathology and Experimental Therapies, School of Medicine, University of Barcelona - UB, 08907 L'Hospitalet de Llobregat, Spain; Immunity, Inflammation, and Cancer Group, Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge - IDIBELL, 08908 L'Hospitalet de Llobregat, Spain
| | - Carmen Bueno-Merino
- Immunology Unit, Department of Pathology and Experimental Therapies, School of Medicine, University of Barcelona - UB, 08907 L'Hospitalet de Llobregat, Spain; Immunity, Inflammation, and Cancer Group, Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge - IDIBELL, 08908 L'Hospitalet de Llobregat, Spain
| | - Ana Del Canizo
- Immunology Unit, Department of Pathology and Experimental Therapies, School of Medicine, University of Barcelona - UB, 08907 L'Hospitalet de Llobregat, Spain; Immunity, Inflammation, and Cancer Group, Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge - IDIBELL, 08908 L'Hospitalet de Llobregat, Spain
| | - Stephanie Riederer
- Division of Virology, Institute for Infection Medicine and Zoonoses, Department of Veterinary Sciences, LMU Munich, 85764 Oberschleiβheim, Germany
| | - Maria Barcia
- Immunology Unit, Department of Pathology and Experimental Therapies, School of Medicine, University of Barcelona - UB, 08907 L'Hospitalet de Llobregat, Spain; Immunity, Inflammation, and Cancer Group, Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge - IDIBELL, 08908 L'Hospitalet de Llobregat, Spain
| | - Katrin Brosinski
- Division of Virology, Institute for Infection Medicine and Zoonoses, Department of Veterinary Sciences, LMU Munich, 85764 Oberschleiβheim, Germany
| | - Michael H Lehmann
- Division of Virology, Institute for Infection Medicine and Zoonoses, Department of Veterinary Sciences, LMU Munich, 85764 Oberschleiβheim, Germany
| | - Asisa Volz
- Division of Virology, Institute for Infection Medicine and Zoonoses, Department of Veterinary Sciences, LMU Munich, 85764 Oberschleiβheim, Germany; Institute of Virology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Xavier Saelens
- Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium; VIB Center for Medical Biotechnology, VIB, 9052 Ghent, Belgium; Department of Biochemistry and Microbiology, Ghent University, 9052 Ghent, Belgium
| | - Gerd Sutter
- Division of Virology, Institute for Infection Medicine and Zoonoses, Department of Veterinary Sciences, LMU Munich, 85764 Oberschleiβheim, Germany; German Center for Infection Research (DZIF), Partner Site Munich, 80539 Munich, Germany
| |
Collapse
|
3
|
Nia GE, Nikpayam E, Farrokhi M, Bolhassani A, Meuwissen R. Advances in cell-based delivery of oncolytic viruses as therapy for lung cancer. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200788. [PMID: 38596310 PMCID: PMC10976516 DOI: 10.1016/j.omton.2024.200788] [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: 04/11/2024]
Abstract
Lung cancer's intractability is enhanced by its frequent resistance to (chemo)therapy and often high relapse rates that make it the leading cause of cancer death worldwide. Improvement of therapy efficacy is a crucial issue that might lead to a significant advance in the treatment of lung cancer. Oncolytic viruses are desirable combination partners in the developing field of cancer immunotherapy due to their direct cytotoxic effects and ability to elicit an immune response. Systemic oncolytic virus administration through intravenous injection should ideally lead to the highest efficacy in oncolytic activity. However, this is often hampered by the prevalence of host-specific, anti-viral immune responses. One way to achieve more efficient systemic oncolytic virus delivery is through better protection against neutralization by several components of the host immune system. Carrier cells, which can even have innate tumor tropism, have shown their appropriateness as effective vehicles for systemic oncolytic virus infection through circumventing restrictive features of the immune system and can warrant oncolytic virus delivery to tumors. In this overview, we summarize promising results from studies in which carrier cells have shown their usefulness for improved systemic oncolytic virus delivery and better oncolytic virus therapy against lung cancer.
Collapse
Affiliation(s)
- Giti Esmail Nia
- Faculty of Allied Medicine, Cellular and Molecular Research Centre, Iran University of Medical Science, Tehran, Iran
- Department of Basic Oncology, Health Institute of Ege University, Izmir, Turkey
| | - Elahe Nikpayam
- Department of Regenerative and Cancer Biology, Albany Medical College, Albany, NY, USA
| | | | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Ralph Meuwissen
- Department of Basic Oncology, Health Institute of Ege University, Izmir, Turkey
- Ege University Translational Pulmonary Research Center (EgeSAM), Ege University, Izmir, Turkey
| |
Collapse
|
4
|
Enow JA, Sheikh HI, Rahman MM. Tumor Tropism of DNA Viruses for Oncolytic Virotherapy. Viruses 2023; 15:2262. [PMID: 38005938 PMCID: PMC10675630 DOI: 10.3390/v15112262] [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: 10/17/2023] [Revised: 11/08/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Oncolytic viruses (OVs) have emerged as one of the most promising cancer immunotherapy agents that selectively target and kill cancer cells while sparing normal cells. OVs are from diverse families of viruses and can possess either a DNA or an RNA genome. These viruses also have either a natural or engineered tropism for cancer cells. Oncolytic DNA viruses have the additional advantage of a stable genome and multiple-transgene insertion capability without compromising infection or replication. Herpes simplex virus 1 (HSV-1), a member of the oncolytic DNA viruses, has been approved for the treatment of cancers. This success with HSV-1 was achievable by introducing multiple genetic modifications within the virus to enhance cancer selectivity and reduce the toxicity to healthy cells. Here, we review the natural characteristics of and genetically engineered changes in selected DNA viruses that enhance the tumor tropism of these oncolytic viruses.
Collapse
Affiliation(s)
- Junior A. Enow
- Biodesign Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Hummad I. Sheikh
- Biodesign Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Masmudur M. Rahman
- Biodesign Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| |
Collapse
|
5
|
Otter AD, Jones S, Hicks B, Bailey D, Callaby H, Houlihan C, Rampling T, Gordon NC, Selman H, Satheshkumar PS, Townsend M, Mehta R, Pond M, Jones R, Wright D, Oeser C, Tonge S, Linley E, Hemingway G, Coleman T, Millward S, Lloyd A, Damon I, Brooks T, Vipond R, Rowe C, Hallis B. Monkeypox virus-infected individuals mount comparable humoral immune responses as Smallpox-vaccinated individuals. Nat Commun 2023; 14:5948. [PMID: 37741831 PMCID: PMC10517934 DOI: 10.1038/s41467-023-41587-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 09/11/2023] [Indexed: 09/25/2023] Open
Abstract
In early 2022, a cluster of monkeypox virus (MPXV) infection (mpox) cases were identified within the UK with no prior travel history to MPXV-endemic regions. Subsequently, case numbers exceeding 80,000 were reported worldwide, primarily affecting gay, bisexual, and other men who have sex with men (GBMSM). Public health agencies worldwide have offered the IMVANEX Smallpox vaccination to these individuals at high-risk to provide protection and limit the spread of MPXV. We have developed a comprehensive array of ELISAs to study poxvirus-induced antibodies, utilising 24 MPXV and 3 Vaccinia virus (VACV) recombinant antigens. Panels of serum samples from individuals with differing Smallpox-vaccine doses and those with prior MPXV infection were tested on these assays, where we observed that one dose of Smallpox vaccination induces a low number of antibodies to a limited number of MPXV antigens but increasing with further vaccination doses. MPXV infection induced similar antibody responses to diverse poxvirus antigens observed in Smallpox-vaccinated individuals. We identify MPXV A27 as a serological marker of MPXV-infection, whilst MPXV M1 (VACV L1) is likely IMVANEX-specific. Here, we demonstrate analogous humoral antigen recognition between both MPXV-infected or Smallpox-vaccinated individuals, with binding to diverse yet core set of poxvirus antigens, providing opportunities for future vaccine (e.g., mRNA) and therapeutic (e.g., mAbs) design.
Collapse
Affiliation(s)
- Ashley D Otter
- Emerging Pathogen Serology group, UK Health Security Agency, Porton Down, Wiltshire, UK.
| | - Scott Jones
- Emerging Pathogen Serology group, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Bethany Hicks
- Emerging Pathogen Serology group, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Daniel Bailey
- Rare and Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Helen Callaby
- Rare and Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Catherine Houlihan
- Rare and Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, Wiltshire, UK
- Department of Infection and Immunity, University College London, London, UK
| | - Tommy Rampling
- Rare and Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, Wiltshire, UK
- The Hospital for Tropical Diseases, University College London Hospital, London, UK
- NIHR University College London Hospitals BRC, London, UK
| | - Nicola Claire Gordon
- Rare and Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Hannah Selman
- Emerging Pathogen Serology group, UK Health Security Agency, Porton Down, Wiltshire, UK
| | | | - Michael Townsend
- Poxvirus and Rabies Branch, Centre for Disease Control and Prevention, Atlanta, GA, USA
| | - Ravi Mehta
- Imperial College Healthcare NHS Trust, London, UK
| | - Marcus Pond
- Imperial College Healthcare NHS Trust, London, UK
| | - Rachael Jones
- Chelsea and Westminster Hospital NHS Foundation Trust, London, UK
| | - Deborah Wright
- Research and Development, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Clarissa Oeser
- Immunisation and Vaccine Preventable Diseases Division, UK Health Security Agency, Colindale, London, UK
| | - Simon Tonge
- Seroepidemiology Unit, UK Health Security Agency, Manchester, UK
| | - Ezra Linley
- Seroepidemiology Unit, UK Health Security Agency, Manchester, UK
| | - Georgia Hemingway
- Emerging Pathogen Serology group, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Tom Coleman
- Emerging Pathogen Serology group, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Sebastian Millward
- Emerging Pathogen Serology group, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Aaron Lloyd
- Emerging Pathogen Serology group, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Inger Damon
- Poxvirus and Rabies Branch, Centre for Disease Control and Prevention, Atlanta, GA, USA
| | - Tim Brooks
- Rare and Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Richard Vipond
- Research and Development, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Cathy Rowe
- Emerging Pathogen Serology group, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Bassam Hallis
- Research and Development, UK Health Security Agency, Porton Down, Wiltshire, UK
| |
Collapse
|
6
|
Li M, Zhang M, Ye Q, Liu Y, Qian W. Preclinical and clinical trials of oncolytic vaccinia virus in cancer immunotherapy: a comprehensive review. Cancer Biol Med 2023; 20:j.issn.2095-3941.2023.0202. [PMID: 37615308 PMCID: PMC10546091 DOI: 10.20892/j.issn.2095-3941.2023.0202] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 07/19/2023] [Indexed: 08/25/2023] Open
Abstract
Oncolytic virotherapy has emerged as a promising treatment for human cancers owing to an ability to elicit curative effects via systemic administration. Tumor cells often create an unfavorable immunosuppressive microenvironment that degrade viral structures and impede viral replication; however, recent studies have established that viruses altered via genetic modifications can serve as effective oncolytic agents to combat hostile tumor environments. Specifically, oncolytic vaccinia virus (OVV) has gained popularity owing to its safety, potential for systemic delivery, and large gene insertion capacity. This review highlights current research on the use of engineered mutated viruses and gene-armed OVVs to reverse the tumor microenvironment and enhance antitumor activity in vitro and in vivo, and provides an overview of ongoing clinical trials and combination therapies. In addition, we discuss the potential benefits and drawbacks of OVV as a cancer therapy, and explore different perspectives in this field.
Collapse
Affiliation(s)
- Mengyuan Li
- Department of Hematology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Minghuan Zhang
- Department of Hematology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Qian Ye
- Hangzhou Rong-Gu Biotechnology Limited Company, Hangzhou 310056, China
| | - Yunhua Liu
- Department of Pathology & Pathophysiology and Department of Surgical Oncology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Wenbin Qian
- Department of Hematology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| |
Collapse
|
7
|
Bécares M, Albert M, Tárrega C, Coloma R, Falqui M, Luhmann EK, Radoshevich L, Guerra S. ISG15 Is Required for the Dissemination of Vaccinia Virus Extracellular Virions. Microbiol Spectr 2023; 11:e0450822. [PMID: 37036376 PMCID: PMC10269806 DOI: 10.1128/spectrum.04508-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 03/15/2023] [Indexed: 04/11/2023] Open
Abstract
Viruses have developed many different strategies to counteract immune responses, and Vaccinia virus (VACV) is one of a kind in this aspect. To ensure an efficient infection, VACV undergoes a complex morphogenetic process resulting in the production of two types of infective virions: intracellular mature virus (MV) and extracellular enveloped virus (EV), whose spread depends on different dissemination mechanisms. MVs disseminate after cell lysis, whereas EVs are released or propelled in actin tails from living cells. Here, we show that ISG15 participates in the control of VACV dissemination. Infection of Isg15-/- mouse embryonic fibroblasts with VACV International Health Department-J (IHD-J) strain resulted in decreased EV production, concomitant with reduced induction of actin tails and the abolition of comet-shaped plaque formation, compared to Isg15+/+ cells. Transmission electron microscopy revealed the accumulation of intracellular virus particles and a decrease in extracellular virus particles in the absence of interferon-stimulated gene 15 (ISG15), a finding consistent with altered virus egress. Immunoblot and quantitative proteomic analysis of sucrose gradient-purified virions from both genotypes reported differences in protein levels and composition of viral proteins present on virions, suggesting an ISG15-mediated control of viral proteome. Lastly, the generation of a recombinant IHD-J expressing V5-tagged ISG15 (IHD-J-ISG15) allowed us to identify several viral proteins as potential ISG15 targets, highlighting the proteins A34 and A36, which are essential for EV formation. Altogether, our results indicate that ISG15 is an important host factor in the regulation of VACV dissemination. IMPORTANCE Viral infections are a constant battle between the virus and the host. While the host's only goal is victory, the main purpose of the virus is to spread and conquer new territories at the expense of the host's resources. Along millions of years of incessant encounters, poxviruses have developed a unique strategy consisting in the production two specialized "troops": intracellular mature virions (MVs) and extracellular virions (EVs). MVs mediate transmission between hosts, and EVs ensure advance on the battlefield mediating the long-range dissemination. The mechanism by which the virus "decides" to shed from the primary site of infection and its significant impact in viral transmission is not yet fully established. Here, we demonstrate that this process is finely regulated by ISG15/ISGylation, an interferon-induced ubiquitin-like protein with broad antiviral activity. Studying the mechanism that viruses use during infection could result in new ways of understanding our perpetual war against disease and how we might win the next great battle.
Collapse
Affiliation(s)
- Martina Bécares
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuel Albert
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Céline Tárrega
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Rocío Coloma
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Michela Falqui
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Emma K. Luhmann
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Lilliana Radoshevich
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Susana Guerra
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
8
|
Zhu X, Fan C, Xiong Z, Chen M, Li Z, Tao T, Liu X. Development and application of oncolytic viruses as the nemesis of tumor cells. Front Microbiol 2023; 14:1188526. [PMID: 37440883 PMCID: PMC10335770 DOI: 10.3389/fmicb.2023.1188526] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/18/2023] [Indexed: 07/15/2023] Open
Abstract
Viruses and tumors are two pathologies that negatively impact human health, but what occurs when a virus encounters a tumor? A global consensus among cancer patients suggests that surgical resection, chemotherapy, radiotherapy, and other methods are the primary means to combat cancer. However, with the innovation and development of biomedical technology, tumor biotherapy (immunotherapy, molecular targeted therapy, gene therapy, oncolytic virus therapy, etc.) has emerged as an alternative treatment for malignant tumors. Oncolytic viruses possess numerous anti-tumor properties, such as directly lysing tumor cells, activating anti-tumor immune responses, and improving the tumor microenvironment. Compared to traditional immunotherapy, oncolytic virus therapy offers advantages including high killing efficiency, precise targeting, and minimal side effects. Although oncolytic virus (OV) therapy was introduced as a novel approach to tumor treatment in the 19th century, its efficacy was suboptimal, limiting its widespread application. However, since the U.S. Food and Drug Administration (FDA) approved the first OV therapy drug, T-VEC, in 2015, interest in OV has grown significantly. In recent years, oncolytic virus therapy has shown increasingly promising application prospects and has become a major research focus in the field of cancer treatment. This article reviews the development, classification, and research progress of oncolytic viruses, as well as their mechanisms of action, therapeutic methods, and routes of administration.
Collapse
Affiliation(s)
- Xiao Zhu
- Zhejiang Provincial People's Hospital Affiliated to Hangzhou Medical College, Hangzhou Medical College, Hangzhou, China
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
- Department of Biological and Chemical Sciences, New York Institute of Technology—Manhattan Campus, New York, NY, United States
| | - Chenyang Fan
- Department of Clinical Medicine, Medicine and Technology, School of Zunyi Medical University, Zunyi, China
| | - Zhuolong Xiong
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Mingwei Chen
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Zesong Li
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital(Shenzhen Institute of Translational Medicine), Shenzhen, China
| | - Tao Tao
- Department of Gastroenterology, Zibo Central Hospital, Zibo, China
| | - Xiuqing Liu
- Department of Clinical Laboratory, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| |
Collapse
|
9
|
Ghabeshi S, Ghasemi S, Mousavizadeh L. The effective factors in human-specific tropism and viral pathogenicity in orthopoxviruses. Cell Biol Int 2023; 47:341-351. [PMID: 36317465 DOI: 10.1002/cbin.11941] [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: 08/07/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 11/08/2022]
Abstract
The orthopoxvirus (OPV) genus includes several species that infect humans, including variola, monkeypox, vaccinia, and cowpox. Variola and monkeypox are often life-threatening diseases, while vaccinia and cowpox are usually associated with local lesions. The epidemic potential for OPVs may be lower than respiratory-borne viruses or RNA viruses. However, OPVs are notable for their spread and distribution in different environments and among different hosts. The emergence or re-emergence of OPVs in the human population can also occur in wild or domestic animals as intermediate hosts. More effective and safer vaccines for poxvirus can be developed by understanding how immunity is regulated in poxvirus and vaccines for DNA viruses. Downstream events in cells affected by the virus are regulated functionally by a series of characteristics that are affected by host cell interactions and responses of cells against viral infections, including the interferon pathway and apoptosis. Furthermore, infection outcome is greatly influenced by the distinct selection of host-range and immune-modulatory genes that confer the potential for pathogenesis and host-to-host transmission and the distinct host-range properties of each immune-modulatory gene. The present study reviewed the effective factors in human-restricted tropism and virus pathogenicity in OPVs.
Collapse
Affiliation(s)
- Soad Ghabeshi
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.,Health Policy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sorayya Ghasemi
- Cancer Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Leila Mousavizadeh
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Zeng J, Li X, Sander M, Zhang H, Yan G, Lin Y. Oncolytic Viro-Immunotherapy: An Emerging Option in the Treatment of Gliomas. Front Immunol 2021; 12:721830. [PMID: 34675919 PMCID: PMC8524046 DOI: 10.3389/fimmu.2021.721830] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 09/16/2021] [Indexed: 01/17/2023] Open
Abstract
The prognosis of malignant gliomas remains poor, with median survival fewer than 20 months and a 5-year survival rate merely 5%. Their primary location in the central nervous system (CNS) and its immunosuppressive environment with little T cell infiltration has rendered cancer therapies mostly ineffective, and breakthrough therapies such as immune checkpoint inhibitors (ICIs) have shown limited benefit. However, tumor immunotherapy is developing rapidly and can help overcome these obstacles. But for now, malignant gliomas remain fatal with short survival and limited therapeutic options. Oncolytic virotherapy (OVT) is a unique antitumor immunotherapy wherein viruses selectively or preferentially kill tumor cells, replicate and spread through tumors while inducing antitumor immune responses. OVTs can also recondition the tumor microenvironment and improve the efficacy of other immunotherapies by escalating the infiltration of immune cells into tumors. Some OVTs can penetrate the blood-brain barrier (BBB) and possess tropism for the CNS, enabling intravenous delivery. Despite the therapeutic potential displayed by oncolytic viruses (OVs), optimizing OVT has proved challenging in clinical development, and marketing approvals for OVTs have been rare. In June 2021 however, as a genetically engineered OV based on herpes simplex virus-1 (G47Δ), teserpaturev got conditional and time-limited approval for the treatment of malignant gliomas in Japan. In this review, we summarize the current state of OVT, the synergistic effect of OVT in combination with other immunotherapies as well as the hurdles to successful clinical use. We also provide some suggestions to overcome the challenges in treating of gliomas.
Collapse
Affiliation(s)
- Jiayi Zeng
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiangxue Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, China
| | - Max Sander
- Department of International Cooperation, Guangzhou Virotech Pharmaceutical Co., Ltd., Guangzhou, China
| | - Haipeng Zhang
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, China
| | - Guangmei Yan
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yuan Lin
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
11
|
Resistance Mechanisms Influencing Oncolytic Virotherapy, a Systematic Analysis. Vaccines (Basel) 2021; 9:vaccines9101166. [PMID: 34696274 PMCID: PMC8537623 DOI: 10.3390/vaccines9101166] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/23/2021] [Accepted: 10/06/2021] [Indexed: 12/15/2022] Open
Abstract
Resistance to therapy is a frequently observed phenomenon in the treatment of cancer, and as with other cancer therapeutics, therapies based on oncolytic viruses also face the challenges of resistance, such as humoral and cellular antiviral responses, and tumor-associated interferon-mediated resistance. In order to identify additional mechanisms of resistance that may contribute to therapeutic failure, we developed a systematic search strategy for studies published in PubMed. We analyzed 6143 articles on oncolytic virotherapy and found that approximately 8% of these articles use resistance terms in the abstract and/or title. Of these 439 articles, 87 were original research. Most of the findings reported pertain to resistance mediated by tumor-cell-dependent interferon signaling. Yet, mechanisms such as epigenetic modifications, hypoxia-mediated inhibition, APOBEC-mediated resistance, virus entry barriers, and spatiotemporal restriction to viral spread, although not frequently assessed, were demonstrated to play a major role in resistance. Similarly, our results suggest that the stromal compartment consisting of, but not limited to, myeloid cells, fibroblasts, and epithelial cells requires more study in relation to therapy resistance using oncolytic viruses. Thus, our findings emphasize the need to assess the stromal compartment and to identify novel mechanisms that play an important role in conferring resistance to oncolytic virotherapy.
Collapse
|
12
|
A potential bat adenovirus-based oncolytic virus targeting canine cancers. Sci Rep 2021; 11:16706. [PMID: 34408176 PMCID: PMC8373906 DOI: 10.1038/s41598-021-96101-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 07/28/2021] [Indexed: 12/17/2022] Open
Abstract
Although a canine adenovirus (CAdV)-based oncolytic virus (OV) candidate targeting canine tumors has been reported, its oncolytic effect could be attenuated by CAdV vaccine-induced neutralizing antibodies in dog patients. To circumvent this issue, we focused on the bat adenovirus (BtAdV) strain, which was previously isolated from healthy microbats. We previously showed that this virus replicated efficiently in canine cell lines and did not serologically cross-react with CAdVs, suggesting that it may offer the possibility of an OV candidate for canine tumors. Here, we tested the growth properties and cytotoxicity of the BtAdV Mm32 strain in a panel of canine tumor cells and found that its characteristics were equivalent to those of CAdVs. To produce an Mm32 construct with enhanced tumor specificity, we established a novel reverse genetics system for BtAdV based on bacterial artificial chromosomes, and generated a recombinant virus, Mm32-E1Ap + cTERTp, by inserting a tumor-specific canine telomerase reverse transcriptase promoter into its E1A regulatory region. The growth and cytotoxicity of this recombinant were superior to those of wild-type Mm32 in canine tumor cells, unlike in normal canine cells. These data suggest that Mm32-E1Ap + cTERTp could be a promising OV for alternative canine cancer therapies.
Collapse
|
13
|
Kurosaki H, Nakatake M, Sakamoto T, Kuwano N, Yamane M, Ishii K, Fujiwara Y, Nakamura T. Anti-Tumor Effects of MAPK-Dependent Tumor-Selective Oncolytic Vaccinia Virus Armed with CD/UPRT against Pancreatic Ductal Adenocarcinoma in Mice. Cells 2021; 10:cells10050985. [PMID: 33922406 PMCID: PMC8145488 DOI: 10.3390/cells10050985] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/14/2021] [Accepted: 04/20/2021] [Indexed: 02/07/2023] Open
Abstract
Engineered vaccinia virus serves as an oncolytic virus for cancer virotherapy. We evaluated the oncolytic characteristics of VGF- and O1-deleted recombinant mitogen-activated protein kinase (MAPK)-dependent vaccinia virus (MDRVV). We found that compared with viruses with the deletion of either gene alone, MDRVV is more attenuated in normal cells and can replicate in cancer cells that exhibit constitutive ERK1/2 activation in the MAPK pathway. We armed MDRVV with a bifunctional fusion gene encoding cytosine deaminase and uracil phosphoribosyltransferase (CD/UPRT), which converts 5-fluorocytosine (5-FC) into chemotherapeutic agents, and evaluated its oncolytic activity alone or in combination with 5-FC in human pancreatic cancer cell lines, tumor mouse models of peritoneal dissemination and liver metastasis, and ex vivo-infected live pancreatic cancer patient-derived tissues. CD/UPRT-armed MDRVV alone could efficiently eliminate pancreatic cancers, and its antitumor effects were partially enhanced in combination with 5-FC in vitro and in vivo. Moreover, the replication of MDRVV was detected in tumor cells of patient-derived, surgically resected tissues, which showed enlarged nuclei and high expression of pERK1/2 and Ki-67, and not in stromal cells. Our findings suggest that systemic injections of CD/UPRT-armed MDRVV alone or in combination with 5-FC are promising therapeutic strategies for pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Hajime Kurosaki
- Division of Molecular Medicine, Department of Genomic Medicine and Regenerative Therapy, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan; (H.K.); (M.N.); (N.K.); (M.Y.); (K.I.)
| | - Motomu Nakatake
- Division of Molecular Medicine, Department of Genomic Medicine and Regenerative Therapy, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan; (H.K.); (M.N.); (N.K.); (M.Y.); (K.I.)
| | - Teruhisa Sakamoto
- Division of Surgical Oncology, Department of Surgery, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan; (T.S.); (Y.F.)
| | - Nozomi Kuwano
- Division of Molecular Medicine, Department of Genomic Medicine and Regenerative Therapy, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan; (H.K.); (M.N.); (N.K.); (M.Y.); (K.I.)
| | - Masato Yamane
- Division of Molecular Medicine, Department of Genomic Medicine and Regenerative Therapy, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan; (H.K.); (M.N.); (N.K.); (M.Y.); (K.I.)
| | - Kenta Ishii
- Division of Molecular Medicine, Department of Genomic Medicine and Regenerative Therapy, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan; (H.K.); (M.N.); (N.K.); (M.Y.); (K.I.)
| | - Yoshiyuki Fujiwara
- Division of Surgical Oncology, Department of Surgery, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan; (T.S.); (Y.F.)
| | - Takafumi Nakamura
- Division of Molecular Medicine, Department of Genomic Medicine and Regenerative Therapy, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan; (H.K.); (M.N.); (N.K.); (M.Y.); (K.I.)
- Correspondence: ; Tel.: +81-859-38-7550; Fax: +81-859-38-6422
| |
Collapse
|
14
|
Wu TM, Liu JB, Liu Y, Shi Y, Li W, Wang GR, Ma YS, Fu D. Power and Promise of Next-Generation Sequencing in Liquid Biopsies and Cancer Control. Cancer Control 2021; 27:1073274820934805. [PMID: 32806937 PMCID: PMC7791471 DOI: 10.1177/1073274820934805] [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] [Indexed: 12/24/2022] Open
Abstract
Traditional methods of cancer treatment are usually based on the morphological
and histological diagnosis of tumors, and they are not optimized according to
the specific situation. Precision medicine adjusts the existing treatment
regimen based on the patient’s genomic information to make it most suitable for
patients. Detection of genetic mutations in tumors is the basis of precise
cancer medicine. Through the analysis of genetic mutations in patients with
cancer, we can tailor the treatment plan for each patient with cancer to
maximize the curative effect, minimize damage to healthy tissues, and optimize
resources. In recent years, next-generation sequencing technology has developed
rapidly and has become the core technology of precise targeted therapy and
immunotherapy for cancer. From early cancer screening to treatment guidance for
patients with advanced cancer, liquid biopsy is increasingly used in cancer
management. This is as a result of the development of better noninvasive,
repeatable, sensitive, and accurate tools used in early screening, diagnosis,
evaluation, and monitoring of patients. Cell-free DNA, which is a new
noninvasive molecular pathological detection method, often carries
tumor-specific gene changes. It plays an important role in optimizing treatment
and evaluating the efficacy of different treatment options in clinical trials,
and it has broad clinical applications.
Collapse
Affiliation(s)
- Ting-Miao Wu
- Department of Radiology, 12485The Fourth Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ji-Bin Liu
- Cancer Institute, 377323Nantong Tumor Hospital, Nantong, China
| | - Yu Liu
- National Engineering Laboratory for Rice and By-product Deep Processing, College of Food Science and Engineering, 12571Central South University of Forestry and Technology, Chaha, China
| | - Yi Shi
- National Engineering Laboratory for Rice and By-product Deep Processing, College of Food Science and Engineering, 12571Central South University of Forestry and Technology, Chaha, China
| | - Wen Li
- National Engineering Laboratory for Rice and By-product Deep Processing, College of Food Science and Engineering, 12571Central South University of Forestry and Technology, Chaha, China
| | - Gao-Ren Wang
- Cancer Institute, 377323Nantong Tumor Hospital, Nantong, China
| | - Yu-Shui Ma
- Cancer Institute, 377323Nantong Tumor Hospital, Nantong, China.,Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, 12476Tongji University School of Medicine, Shanghai, China
| | - Da Fu
- Department of Radiology, 12485The Fourth Affiliated Hospital of Anhui Medical University, Hefei, China.,Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, 12476Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
15
|
Zhang Z, Dong L, Zhao C, Zheng P, Zhang X, Xu J. Vaccinia virus-based vector against infectious diseases and tumors. Hum Vaccin Immunother 2021; 17:1578-1585. [PMID: 33606578 DOI: 10.1080/21645515.2020.1840887] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Vaccinia virus was used to prevent smallpox. After the World Health Organization declared smallpox extinct, vaccinia virus has been explored for the development of vaccines against a variety of infectious diseases. It also finds a new place in oncolytic therapy. Here we provide a brief review of the history, current status, and future prospect of vaccinia virus-based vaccine and oncolytic virus. New advancements, including a single vaccine targeting multiple viruses, strategies of arming vaccinia viruses to enhance anti-tumor activity, the promise and challenge of combining vaccinia-based virotherapy with immunotherapy, are discussed as special focus.
Collapse
Affiliation(s)
- Ziling Zhang
- Shanghai Public Health ClinicalCenter& Institutes of Biomedical Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lanlan Dong
- Shanghai Public Health ClinicalCenter& Institutes of Biomedical Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chen Zhao
- Shanghai Public Health ClinicalCenter& Institutes of Biomedical Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Peiyong Zheng
- Institute of Digestive Diseases, LongHua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoyan Zhang
- Shanghai Public Health ClinicalCenter& Institutes of Biomedical Science, Shanghai Medical College, Fudan University, Shanghai, China.,State Key Laboratory for Infectious Disease Prevention and Control, China Centers for Disease Control and Prevention, Beijing, China
| | - Jianqing Xu
- Shanghai Public Health ClinicalCenter& Institutes of Biomedical Science, Shanghai Medical College, Fudan University, Shanghai, China.,State Key Laboratory for Infectious Disease Prevention and Control, China Centers for Disease Control and Prevention, Beijing, China
| |
Collapse
|
16
|
Oncolytic vaccinia virus induces a novel phenotype of CD8 + effector T cells characterized by high ICOS expression. MOLECULAR THERAPY-ONCOLYTICS 2021; 20:422-432. [PMID: 33665362 PMCID: PMC7900640 DOI: 10.1016/j.omto.2021.01.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 01/26/2021] [Indexed: 12/21/2022]
Abstract
Characterization of the intratumoral immune status is important for developing immunotherapies and evaluating their antitumor effectiveness. CD8+ T cells are one of the most important cell types that directly and indirectly contribute to antitumor efficacy by releasing cytolytic molecules and inflammatory cytokines in the tumor microenvironment. Previously, we engineered a tumor-selective oncolytic vaccinia virus that encodes interleukin-7 (IL-7) and IL-12 and demonstrated its usefulness as an agent for in situ vaccination against tumors, with data showing that antitumor efficacy was reliant upon CD8+ T cells recruited by viral treatment. Here, we investigated the phenotypic changes in intratumoral CD8+ T cells caused by this oncolytic virus and found increased expression of inducible co-stimulator (ICOS) in PD-1-CD8+ T cells. Unlike previously reported ICOS+CD8+ T cells, a subset of ICOS+PD-1-CD8+ T cells showed effector function characterized by granzyme B expression. ICOS expression was induced by the backbone virus, which did not encode any immune transgenes and was independent of upregulation of the type I interferon pathway. Not only did we identify a novel effector cell subset characterized by ICOS expression, but our findings also shed light on a potential unknown aspect of the mechanism of oncolytic vaccinia virotherapy.
Collapse
|
17
|
Fusogenic oncolytic vaccinia virus enhances systemic antitumor immune response by modulating the tumor microenvironment. Mol Ther 2020; 29:1782-1793. [PMID: 33348052 DOI: 10.1016/j.ymthe.2020.12.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/04/2020] [Accepted: 12/15/2020] [Indexed: 11/23/2022] Open
Abstract
Oncolytic viruses induce antitumor immunity following direct viral oncolysis. However, their therapeutic effects are limited in distant untreated tumors because their antitumor function depends on indirect antitumor immunity. Here, we generated a novel fusogenic oncolytic vaccinia virus (FUVAC) and compared its antitumor activity with that of its parental non-fusogenic virus. Compared with the parent, FUVAC exerted the cytopathic effect and induced immunogenic cell death in human and murine cancer cells more efficiently. In a bilateral tumor-bearing syngeneic mouse model, FUVAC administration significantly inhibited tumor growth in both treated and untreated tumors. However, its antitumor effects were completely suppressed by CD8+ T cell depletion. Notably, FUVAC reduced the number of tumor-associated immune-suppressive cells in treated tumors, but not in untreated tumors. Mice treated with FUVAC before an immune checkpoint inhibitor (ICI) treatment achieved complete response (CR) in both treated and untreated tumors, whereas ICI alone did not show antitumor activity. Mice achieving CR rejected rechallenge with the same tumor cells, suggesting establishment of a long-term tumor-specific immune memory. Thus, FUVAC improves the tumor immune microenvironment and enhances systemic antitumor immunity, suggesting that, alone and in combination with ICI, it is a novel immune modulator for overcoming oncolytic virus-resistant tumors.
Collapse
|
18
|
Shin J, Hong SO, Kim M, Lee H, Choi H, Kim J, Hong J, Kang H, Lee E, Lee S, Kong B, Kim M, Choi H, Kim S. Generation of a Novel Oncolytic Vaccinia Virus Using the IHD-W Strain. Hum Gene Ther 2020; 32:517-527. [PMID: 32854548 PMCID: PMC8140350 DOI: 10.1089/hum.2020.050] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Oncolytic viruses are promising cancer therapies due to their selective killing of tumor cells and ability to stimulate the host immune system. As an oncolytic virus platform, vaccinia virus has unique advantages, including rapid replication, a broad range of host targets, and a large capacity for transgene incorporation. In this study, we developed a novel oncolytic vaccinia virus with high potency and a favorable safety profile. We began with the International Health Department-White (IHD-W) strain, which had the strongest cytotoxicity against tumor cells among the four vaccinia virus strains tested. Next, several candidate viruses were constructed by deleting three viral genes (C11R, K3L, and J2R) in various combinations, and their efficacy and safety were compared. The virus ultimately selected, named KLS-3010, exhibited strong antitumor activity against broad targets in vitro and in vivo. Furthermore, KLS-3010 showed a favorable safety profile in mice, as determined by the biodistribution and body weight change. More promisingly, KLS-3010 was able to shift the tumor microenvironment to a proinflammatory state, as evidenced by an increase in activated lymphocytes after KLS-3010 administration, suggesting that this strain may elicit an oncolytic virus-mediated immune response. The KLS-3010 strain thus represents a promising platform for the further development of oncolytic virus-based cancer therapies.
Collapse
Affiliation(s)
- Jaeil Shin
- Institute of BioInnovation Research, Kolon Life Science, Seoul, Republic of Korea
| | - Soon-Oh Hong
- Institute of BioInnovation Research, Kolon Life Science, Seoul, Republic of Korea
| | - Minju Kim
- Institute of BioInnovation Research, Kolon Life Science, Seoul, Republic of Korea
| | - Hyesun Lee
- Institute of BioInnovation Research, Kolon Life Science, Seoul, Republic of Korea
| | - Hwanjun Choi
- Institute of BioInnovation Research, Kolon Life Science, Seoul, Republic of Korea
| | - Joonsung Kim
- Institute of BioInnovation Research, Kolon Life Science, Seoul, Republic of Korea
| | - Jieun Hong
- Institute of BioInnovation Research, Kolon Life Science, Seoul, Republic of Korea
| | - Hyesoo Kang
- Institute of BioInnovation Research, Kolon Life Science, Seoul, Republic of Korea
| | - Eunjin Lee
- Institute of BioInnovation Research, Kolon Life Science, Seoul, Republic of Korea
| | - Soondong Lee
- Institute of BioInnovation Research, Kolon Life Science, Seoul, Republic of Korea
| | - Byoungjae Kong
- Institute of BioInnovation Research, Kolon Life Science, Seoul, Republic of Korea
| | - Minjung Kim
- Institute of BioInnovation Research, Kolon Life Science, Seoul, Republic of Korea
| | - Heonsik Choi
- Institute of BioInnovation Research, Kolon Life Science, Seoul, Republic of Korea
| | - Sujeong Kim
- Institute of BioInnovation Research, Kolon Life Science, Seoul, Republic of Korea
| |
Collapse
|
19
|
Abstract
Therapeutic viral gene delivery is an emerging technology which aims to correct genetic mutations by introducing new genetic information to cells either to correct a faulty gene or to initiate cell death in oncolytic treatments. In recent years, significant scientific progress has led to several clinical trials resulting in the approval of gene therapies for human treatment. However, successful therapies remain limited due to a number of challenges such as inefficient cell uptake, low transduction efficiency (TE), limited tropism, liver toxicity and immune response. To adress these issues and increase the number of available therapies, additives from a broad range of materials like polymers, peptides, lipids, nanoparticles, and small molecules have been applied so far. The scope of this review is to highlight these selected delivery systems from a materials perspective.
Collapse
Affiliation(s)
- Kübra Kaygisiz
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | | |
Collapse
|