1
|
Sun L, Zhao Q, Miao L. Combination therapy with oncolytic viruses for lung cancer treatment. Front Oncol 2025; 15:1524079. [PMID: 40248194 PMCID: PMC12003109 DOI: 10.3389/fonc.2025.1524079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 03/18/2025] [Indexed: 04/19/2025] Open
Abstract
Lung cancer is the leading cause of cancer-related death globally. Despite various treatment options, adverse reactions and treatment resistance limit their clinical application and efficacy, therefore, new effective treatment options are still needed. Oncolytic viruses (OVs) are a new anti-cancer option. With a powerful anti-tumor effect, OVs are gradually being applied to the treatment of solid tumor. In clinical practice, we have found that in patients with NSCLC and SCLC, OVs combined with immune checkpoint inhibitors (ICI) treatment make tumor with poor response to immunotherapy become sensitive. Furthermore, studies have shown that OVs combined with chemotherapy, radiation therapy, and other immune approaches (such as anti-pd1 drugs) have synergistic effects. These studies suggest that OVs combined therapy may bring hope for the treatment of lung cancer patients. This article will review the current status and prospect of OVs combination therapy in the field of lung cancer treatment and summarizes the mechanism of action.
Collapse
Affiliation(s)
- Lei Sun
- Department of Pharmacy, Yancheng Branch of Nanjing Drum Tower Hospital, Yancheng, Jiangsu, China
| | - Qi Zhao
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Nanjing, Jiangsu, China
| | - Liyun Miao
- Department of Pharmacy, Yancheng Branch of Nanjing Drum Tower Hospital, Yancheng, Jiangsu, China
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Nanjing, Jiangsu, China
| |
Collapse
|
2
|
Shakiba Y, Rahman MM. Testing Oncolytic Myxoma Virus in Immunocompetent Mouse Model for Cancer Therapy. Methods Mol Biol 2025; 2860:353-360. [PMID: 39621278 PMCID: PMC11724751 DOI: 10.1007/978-1-0716-4160-6_22] [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] [Indexed: 01/13/2025]
Abstract
Oncolytic viruses (OVs) have emerged as a class of novel cancer immunotherapeutic. Members of both DNA and RNA viruses developed as OVs for treating diverse types of human cancers. Preclinical research assessing immunotherapeutic efficacy is an essential step toward further development of these OVs. Mice tumor model systems are widely used in preclinical oncolytic viral therapies for evaluating the treatment regimens' efficacy. However, choosing the most appropriate model for a study can be challenging. Here, we describe a simple method of establishing subcutaneous tumors in immunocompetent mice, intratumoral injection of the virus, measuring tumor burden, and studying the survival of mice for preclinical development of oncolytic myxoma virus (MYXV).
Collapse
Affiliation(s)
- Yasmin Shakiba
- School of Medical and Biological Physics, Moscow Institute of Physics and Technology, Moscow, Russia
| | - Masmudur M Rahman
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ, USA.
- School of Life Sciences, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
3
|
Zhang Y, Shi X, Shen Y, Dong X, He R, Chen G, Zhang Y, Tan H, Zhang K. Nanoengineering-armed oncolytic viruses drive antitumor response: progress and challenges. MedComm (Beijing) 2024; 5:e755. [PMID: 39399642 PMCID: PMC11467370 DOI: 10.1002/mco2.755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/01/2024] [Accepted: 09/03/2024] [Indexed: 10/15/2024] Open
Abstract
Oncolytic viruses (OVs) have emerged as a powerful tool in cancer therapy. Characterized with the unique abilities to selectively target and lyse tumor cells, OVs can expedite the induction of cell death, thereby facilitating effective tumor eradication. Nanoengineering-derived OVs overcome traditional OV therapy limitations by enhancing the stability of viral circulation, and tumor targeting, promising improved clinical safety and efficacy and so on. This review provides a comprehensive analysis of the multifaceted mechanisms through which engineered OVs can suppress tumor progression. It initiates with a concise delineation on the fundamental attributes of existing OVs, followed by the exploration of their mechanisms of the antitumor response. Amid rapid advancements in nanomedicine, this review presents an extensive overview of the latest developments in the synergy between nanomaterials, nanotechnologies, and OVs, highlighting the unique characteristics and properties of the nanomaterials employed and their potential to spur innovation in novel virus design. Additionally, it delves into the current challenges in this emerging field and proposes strategies to overcome these obstacles, aiming to spur innovation in the design and application of next-generation OVs.
Collapse
Affiliation(s)
- Yan Zhang
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
- Department of VIP ClinicGeneral Division, Shanghai East Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Xinyu Shi
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
- Department of VIP ClinicGeneral Division, Shanghai East Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Yifan Shen
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
- Department of VIP ClinicGeneral Division, Shanghai East Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Xiulin Dong
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Ruiqing He
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Guo Chen
- Department of VIP ClinicGeneral Division, Shanghai East Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Yan Zhang
- Department of Medical UltrasoundRenji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Honghong Tan
- Department of VIP ClinicGeneral Division, Shanghai East Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Kun Zhang
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| |
Collapse
|
4
|
Franks ML, An JH, Leavenworth JW. The Role of Natural Killer Cells in Oncolytic Virotherapy: Friends or Foes? Vaccines (Basel) 2024; 12:721. [PMID: 39066359 PMCID: PMC11281503 DOI: 10.3390/vaccines12070721] [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: 05/31/2024] [Revised: 06/26/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
Oncolytic virotherapy (OVT) has emerged as a promising cancer immunotherapy, and is capable of potentiating other immunotherapies due to its capacity to increase tumor immunogenicity and to boost host antitumor immunity. Natural killer (NK) cells are a critical cellular component for mediating the antitumor response, but hold a mixed reputation for their role in mediating the therapeutic efficacy of OVT. This review will discuss the pros and cons of how NK cells impact OVT, and how to harness this knowledge for the development of effective strategies that could modulate NK cells to improve OVT-based therapeutic outcomes.
Collapse
Affiliation(s)
- Michael L. Franks
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (M.L.F.)
- Graduate Biomedical Sciences Program, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ju-Hyun An
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (M.L.F.)
| | - Jianmei W. Leavenworth
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (M.L.F.)
- The O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
5
|
Nie L, Huang Y, Cheng Z, Luo H, Zhan Y, Dou K, Ma C, Yu C, Luo C, Liu Z, Liu S, Zhu Y. An intranasal influenza virus vector vaccine protects against Helicobacter pylori in mice. J Virol 2024; 98:e0192323. [PMID: 38358289 PMCID: PMC10949480 DOI: 10.1128/jvi.01923-23] [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: 12/09/2023] [Accepted: 01/22/2024] [Indexed: 02/16/2024] Open
Abstract
Helicobacter pylori is a human pathogen that infects almost half of the population. Antibiotic resistance in H. pylori threatens health and increases the demand for prophylactic and therapeutic vaccines. Traditional oral vaccine research faces considerable challenges because of the epithelial barrier, potential enterotoxicity of adjuvants, and the challenging conditions of the gastric environment. We developed an intranasal influenza A virus (IAV) vector vaccine based on two live attenuated influenza viruses with modified acidic polymerase protein (PA) genes encoding the A subunit of H. pylori neutrophil-activating protein (NapA), named IAV-NapA, including influenza virus A/WSN/33 (WSN)-NapA and A/Puerto Rico/8/34 (PR8)-NapA. These recombinant influenza viruses were highly attenuated and exhibited strong immunogenicity in mice. Vaccination with IAV-NapA induced antigen-specific humoral and mucosal immune responses while stimulating robust Th1 and Th17 cell immune responses in mice. Our findings suggest that prophylactic and therapeutic vaccination with influenza virus vector vaccines significantly reduces colonization of H. pylori and inflammation in the stomach of mice.IMPORTANCEHelicobacter pylori is the most common cause of chronic gastritis and leads to severe gastroduodenal pathology in some patients. Many studies have shown that Th1 and Th17 cellular and gastric mucosal immune responses are critical in reducing H. pylori load. IAV vector vaccines can stimulate these immune responses while overcoming potential adjuvant toxicity and antigen dosing issues. To date, no studies have demonstrated the role of live attenuated IAV vector vaccines in preventing and treating H. pylori infection. Our work indicates that vaccination with IAV-NapA induces antigen-specific humoral, cellular, and mucosal immunity, producing a protective and therapeutic effect against H. pylori infection in BALB/c mice. This undescribed H. pylori vaccination approach may provide valuable information for developing vaccines against H. pylori infection.
Collapse
Affiliation(s)
- Longyu Nie
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yu Huang
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Zhikui Cheng
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Hao Luo
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yuxin Zhan
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Kaiwen Dou
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Caijiao Ma
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Chen Yu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Chuanjin Luo
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Zhiqiang Liu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Shi Liu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Ying Zhu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
6
|
Ji D, Zhang Y, Sun J, Zhang B, Ma W, Cheng B, Wang X, Li Y, Mu Y, Xu H, Wang Q, Zhang C, Xiao S, Zhang L, Zhou D. An engineered influenza virus to deliver antigens for lung cancer vaccination. Nat Biotechnol 2024; 42:518-528. [PMID: 37231262 DOI: 10.1038/s41587-023-01796-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 04/20/2023] [Indexed: 05/27/2023]
Abstract
The development of cancer neoantigen vaccines that prime the anti-tumor immune responses has been hindered in part by challenges in delivery of neoantigens to the tumor. Here, using the model antigen ovalbumin (OVA) in a melanoma model, we demonstrate a chimeric antigenic peptide influenza virus (CAP-Flu) system for delivery of antigenic peptides bound to influenza A virus (IAV) to the lung. We conjugated attenuated IAVs with the innate immunostimulatory agent CpG and, after intranasal administration to the mouse lung, observed increased immune cell infiltration to the tumor. OVA was then covalently displayed on IAV-CPG using click chemistry. Vaccination with this construct yielded robust antigen uptake by dendritic cells, a specific immune cell response and a significant increase in tumor-infiltrating lymphocytes compared to peptides alone. Lastly, we engineered the IAV to express anti-PD1-L1 nanobodies that further enhanced regression of lung metastases and prolonged mouse survival after rechallenge. Engineered IAVs can be equipped with any tumor neoantigen of interest to generate lung cancer vaccines.
Collapse
Affiliation(s)
- Dezhong Ji
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China.
- Shenzhen Bay Laboratory, Gaoke International Innovation Center, Shenzhen, China.
- Peking University Ningbo Institute of Marine Medicines, Ningbo, China.
| | - Yuanjie Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Shenzhen Bay Laboratory, Gaoke International Innovation Center, Shenzhen, China
- Peking University Ningbo Institute of Marine Medicines, Ningbo, China
| | - Jiaqi Sun
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Shenzhen Bay Laboratory, Gaoke International Innovation Center, Shenzhen, China
- Peking University Ningbo Institute of Marine Medicines, Ningbo, China
| | - Bo Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Wenxiao Ma
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Shenzhen Bay Laboratory, Gaoke International Innovation Center, Shenzhen, China
- Peking University Ningbo Institute of Marine Medicines, Ningbo, China
| | - Boyang Cheng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Shenzhen Bay Laboratory, Gaoke International Innovation Center, Shenzhen, China
- Peking University Ningbo Institute of Marine Medicines, Ningbo, China
| | - Xinchen Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Shenzhen Bay Laboratory, Gaoke International Innovation Center, Shenzhen, China
- Peking University Ningbo Institute of Marine Medicines, Ningbo, China
| | - Yuanhao Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Shenzhen Bay Laboratory, Gaoke International Innovation Center, Shenzhen, China
- Peking University Ningbo Institute of Marine Medicines, Ningbo, China
| | - Yu Mu
- Shenzhen Bay Laboratory, Gaoke International Innovation Center, Shenzhen, China
| | - Huan Xu
- Shenzhen Bay Laboratory, Gaoke International Innovation Center, Shenzhen, China
| | - Qi Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Chuanling Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Peking University Ningbo Institute of Marine Medicines, Ningbo, China
| | - Sulong Xiao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Peking University Ningbo Institute of Marine Medicines, Ningbo, China
| | - Lihe Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Peking University Ningbo Institute of Marine Medicines, Ningbo, China
| | - Demin Zhou
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China.
- Shenzhen Bay Laboratory, Gaoke International Innovation Center, Shenzhen, China.
- Peking University Ningbo Institute of Marine Medicines, Ningbo, China.
| |
Collapse
|
7
|
Ogasawara S. Replication-competent influenza virus with a protein-responsive multiplication ability. N Biotechnol 2023; 77:100-110. [PMID: 37586547 DOI: 10.1016/j.nbt.2023.08.001] [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: 02/26/2023] [Revised: 07/30/2023] [Accepted: 08/12/2023] [Indexed: 08/18/2023]
Abstract
Applications of influenza A viruses (IAV) for virotherapy and biotechnology have accelerated substantially with the development of reverse genetic technology and advances in the understanding of packaging signals. While the use of a replication-competent IAV is particularly promising, owing to its efficient transmission to organ depths with high infectivity, there is also a risk that its multiplication cannot be controlled in a cell-type-specific manner, causing an infectious disease. Therefore, here a simple and effective replication-competent IAV-based cell-targeting system has been developed. It was demonstrated that the activity of the ribonucleoprotein complex (RNP) of IAV could be regulated by the interaction between the endogenous protein and a nanobody fused to the subunit of RNA-dependent RNA polymerase (RdRp). To validate the feasibility of the method, it was demonstrated that RNP containing RdRp fused with Nb139, a nanobody against p53, is inactive in HEK293T cells expressing endogenous p53, but active in p53-defective Saos-2 cells. Finally, a replication-competent IAV was successfully generated that multiplies only in p53-defective tumor cells and an IAV vector was developed that can deliver a foreign gene in cell type-specific manner. The method is flexible because the nanobody can be easily altered to target a different cell type, offering a valuable platform for virotherapy and biotechnology.
Collapse
Affiliation(s)
- Shinzi Ogasawara
- Department of Biology, Faculty of Science, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan.
| |
Collapse
|
8
|
Wong B, Birtch R, Rezaei R, Jamieson T, Crupi MJF, Diallo JS, Ilkow CS. Optimal delivery of RNA interference by viral vectors for cancer therapy. Mol Ther 2023; 31:3127-3145. [PMID: 37735876 PMCID: PMC10638062 DOI: 10.1016/j.ymthe.2023.09.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/28/2023] [Accepted: 09/14/2023] [Indexed: 09/23/2023] Open
Abstract
In recent years, there has been a surge in the innovative modification and application of the viral vector-based gene therapy field. Significant and consistent improvements in the engineering, delivery, and safety of viral vectors have set the stage for their application as RNA interference (RNAi) delivery tools. Viral vector-based delivery of RNAi has made remarkable breakthroughs in the treatment of several debilitating diseases and disorders (e.g., neurological diseases); however, their novelty has yet to be fully applied and utilized for the treatment of cancer. This review highlights the most promising and emerging viral vector delivery tools for RNAi therapeutics while discussing the variables limiting their success and suitability for cancer therapy. Specifically, we outline different integrating and non-integrating viral platforms used for gene delivery, currently employed RNAi targets for anti-cancer effect, and various strategies used to optimize the safety and efficacy of these RNAi therapeutics. Most importantly, we provide great insight into what challenges exist in their application as cancer therapeutics and how these challenges can be effectively navigated to advance the field.
Collapse
Affiliation(s)
- Boaz Wong
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Rayanna Birtch
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Reza Rezaei
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Taylor Jamieson
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Mathieu J F Crupi
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Jean-Simon Diallo
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Carolina S Ilkow
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
9
|
Lovatt C, Parker AL. Oncolytic Viruses and Immune Checkpoint Inhibitors: The "Hot" New Power Couple. Cancers (Basel) 2023; 15:4178. [PMID: 37627206 PMCID: PMC10453115 DOI: 10.3390/cancers15164178] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/15/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer care and shown remarkable efficacy clinically. This efficacy is, however, limited to subsets of patients with significant infiltration of lymphocytes into the tumour microenvironment. To extend their efficacy to patients who fail to respond or achieve durable responses, it is now becoming evident that complex combinations of immunomodulatory agents may be required to extend efficacy to patients with immunologically "cold" tumours. Oncolytic viruses (OVs) have the capacity to selectively replicate within and kill tumour cells, resulting in the induction of immunogenic cell death and the augmentation of anti-tumour immunity, and have emerged as a promising modality for combination therapy to overcome the limitations seen with ICIs. Pre-clinical and clinical data have demonstrated that OVs can increase immune cell infiltration into the tumour and induce anti-tumour immunity, thus changing a "cold" tumour microenvironment that is commonly associated with poor response to ICIs, to a "hot" microenvironment which can render patients more susceptible to ICIs. Here, we review the major viral vector platforms used in OV clinical trials, their success when used as a monotherapy and when combined with adjuvant ICIs, as well as pre-clinical studies looking at the effectiveness of encoding OVs to deliver ICIs locally to the tumour microenvironment through transgene expression.
Collapse
Affiliation(s)
- Charlotte Lovatt
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK;
| | - Alan L. Parker
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK;
- Systems Immunity University Research Institute, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| |
Collapse
|
10
|
Wan PKT, Fernandes RA, Seymour LW. Oncolytic viruses and antibodies: are they more successful when delivered separately or when engineered as a single agent? J Immunother Cancer 2023; 11:e006518. [PMID: 37541690 PMCID: PMC10407364 DOI: 10.1136/jitc-2022-006518] [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] [Accepted: 06/25/2023] [Indexed: 08/06/2023] Open
Abstract
Oncolytic viruses (OVs) provide the promise of tumor-selective cytotoxicity coupled with amplification of the therapeutic agent (the virus) in situ within the tumor improving its therapeutic index. Despite this promise, however, single agent-treatments have not been as successful as combination therapies, particularly combining with checkpoint inhibitor antibodies. The antibodies may be delivered by two approaches, either encoded within the OV genome to restrict antibody production to sites of active virus infection or alternatively given alongside OVs as separate treatments. Both approaches have shown promising therapeutic outcomes, and this leads to an interesting question of whether one approach is potentially better than the other. In this review, we provide a brief summary of the combination OV-antibody therapies that target tumor cells, tumor microenvironment and immune cells to help define key parameters influencing which approach is superior, thereby improving insight into the rational design of OV treatment strategies.
Collapse
|
11
|
You L, Liu Y, Chen N, Zhu L, Xu G, Lv Z, Zhou Y, Li C, Tong H, Jin J, Meng H. Influenza A (H1N1) virus induced long-term remission in a refractory acute myeloid leukaemia. Br J Haematol 2023; 202:745-748. [PMID: 37230766 DOI: 10.1111/bjh.18876] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 05/01/2023] [Accepted: 05/09/2023] [Indexed: 05/27/2023]
Abstract
There have been reports of haematological cancer patients achieving spontaneous remission after being infected with the influenza A or SARS-COV-2 virus. Here, we present the first case of long-term complete remission (CR) induced by influenza A (IAV, H1N1 subtype) in a refractory AML patient and have functionally validated this finding in two different animal disease models. We observed a significant increase in the proportion of helper T cells in the patient after IAV infection. The levels of cytokines, including IL-2, IL-4, IL-6, IL-10, IL-17A, IFN-γ and TNF-α, were higher in IAV-infected patients compared with control groups. These findings indicate that the anti-tumour effects induced by IAV are closely related to the modification of the immune response. Our study provides new evidence of the anti-tumour effects of IAV from a clinical practice perspective.
Collapse
Affiliation(s)
- Liangshun You
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
- Zhejiang Province Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou, Zhejiang, P.R. China
| | - Yi Liu
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Nianci Chen
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Li Zhu
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Gaixiang Xu
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
- Zhejiang Province Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou, Zhejiang, P.R. China
| | - Zuopo Lv
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Yile Zhou
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
- Zhejiang Province Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou, Zhejiang, P.R. China
| | - Chenying Li
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
- Zhejiang Province Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou, Zhejiang, P.R. China
| | - Hongyan Tong
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
- Zhejiang Province Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou, Zhejiang, P.R. China
| | - Jie Jin
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
- Zhejiang Province Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou, Zhejiang, P.R. China
| | - Haitao Meng
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
- Zhejiang Province Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou, Zhejiang, P.R. China
| |
Collapse
|
12
|
Sun F, Xu Y, Deng Z, Yang P. A recombinant oncolytic influenza virus expressing a PD-L1 antibody induces CD8 + T-cell activation via the cGas-STING pathway in mice with hepatocellular carcinoma. Int Immunopharmacol 2023; 120:110323. [PMID: 37207446 DOI: 10.1016/j.intimp.2023.110323] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/21/2023] [Accepted: 05/08/2023] [Indexed: 05/21/2023]
Abstract
OBJECTIVE To evaluate targeted killing of hepatocellular carcinoma (HCC) cells by a recombinant oncolytic influenza virus expressing a PD-L1 antibody (rgFlu/PD-L1) and to develop a novel immunotherapy for HCC. METHODS Using influenza virus reverse genetics, a recombinant oncolytic virus was generated in the background of the A/Puerto Rico/8/34 (PR8) virus, then identified via screening and passage in specific pathogen-free chicken embryos. Hepatocellular carcinoma cell killing by rgFlu/PD-L1 was confirmed in vitro and in vivo. Transcriptome analyses were used to explore PD-L1 expression and function. Western blotting revealed that PD-L1 activated the cGas-STING pathway. RESULTS rgFlu/PD-L1 expressed the PD-L1 heavy and light chain in PB1 and PA, respectively; PR8 served as the backbone. The hemagglutinin titer of rgFlu/PD-L1 was 29, and the virus titer was 9-10 logTCID50/mL. Electron microscopy revealed that the rgFlu/PD-L1 morphology and size were consistent with wild-type influenza virus. The MTS assay showed that rgFlu/PD-L1 induced significant killing of HCC cells but not normal cells. rgFlu/PD-L1 inhibited PD-L1 expression and induced apoptosis in HepG2 cells. Notably, rgFlu/PD-L1 controlled the viability and function of CD8+ T cells by activating the cGas-STING pathway. CONCLUSION rgFlu/PD-L1 activated the cGas-STING pathway in CD8+ T cells, causing them to kill HCC cells. This approach represents a novel immunotherapy for liver cancer.
Collapse
Affiliation(s)
- Fang Sun
- Faculty of Hepato-Pancreato-Biliary Surgery, the First Medical Centre, Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of Chinese PLA, Key Laboratory of Digital Hepatobiliary Surgery, PLA, Beijing, China; National Clinical Research Center for Infectious Diseases, the Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Yan Xu
- Faculty of Hepato-Pancreato-Biliary Surgery, the First Medical Centre, Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of Chinese PLA, Key Laboratory of Digital Hepatobiliary Surgery, PLA, Beijing, China
| | - Zhuoya Deng
- Faculty of Hepato-Pancreato-Biliary Surgery, the First Medical Centre, Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of Chinese PLA, Key Laboratory of Digital Hepatobiliary Surgery, PLA, Beijing, China
| | - Penghui Yang
- Faculty of Hepato-Pancreato-Biliary Surgery, the First Medical Centre, Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of Chinese PLA, Key Laboratory of Digital Hepatobiliary Surgery, PLA, Beijing, China.
| |
Collapse
|
13
|
Awad RM, Breckpot K. Novel technologies for applying immune checkpoint blockers. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 382:1-101. [PMID: 38225100 DOI: 10.1016/bs.ircmb.2023.03.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
Cancer cells develop several ways to subdue the immune system among others via upregulation of inhibitory immune checkpoint (ICP) proteins. These ICPs paralyze immune effector cells and thereby enable unfettered tumor growth. Monoclonal antibodies (mAbs) that block ICPs can prevent immune exhaustion. Due to their outstanding effects, mAbs revolutionized the field of cancer immunotherapy. However, current ICP therapy regimens suffer from issues related to systemic administration of mAbs, including the onset of immune related adverse events, poor pharmacokinetics, limited tumor accessibility and immunogenicity. These drawbacks and new insights on spatiality prompted the exploration of novel administration routes for mAbs for instance peritumoral delivery. Moreover, novel ICP drug classes that are adept to novel delivery technologies were developed to circumvent the drawbacks of mAbs. We therefore review the state-of-the-art and novel delivery strategies of ICP drugs.
Collapse
Affiliation(s)
- Robin Maximilian Awad
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
14
|
Du R, Cui Q, Chen Z, Zhao X, Lin X, Rong L. Revisiting influenza A virus life cycle from a perspective of genome balance. Virol Sin 2023; 38:1-8. [PMID: 36309307 PMCID: PMC10006207 DOI: 10.1016/j.virs.2022.10.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 10/18/2022] [Indexed: 11/06/2022] Open
Abstract
Influenza A virus (IAV) genome comprises eight negative-sense RNA segments, of which the replication is well orchestrated and the delicate balance of multiple segments are dynamically regulated throughout IAV life cycle. However, previous studies seldom discuss these balances except for functional hemagglutinin-neuraminidase balance that is pivotal for both virus entry and release. Therefore, we attempt to revisit IAV life cycle by highlighting the critical role of "genome balance". Moreover, we raise a "balance regression" model of IAV evolution that the virus evolves to rebalance its genome after reassortment or interspecies transmission, and direct a "balance compensation" strategy to rectify the "genome imbalance" as a result of artificial modifications during creation of recombinant IAVs. This review not only improves our understanding of IAV life cycle, but also facilitates both basic and applied research of IAV in future.
Collapse
Affiliation(s)
- Ruikun Du
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China; Qingdao Academy of Chinese Medicinal Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, 266122, China.
| | - Qinghua Cui
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China; Qingdao Academy of Chinese Medicinal Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, 266122, China
| | - Zinuo Chen
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Xiujuan Zhao
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Xiaojing Lin
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Lijun Rong
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, 60612, USA.
| |
Collapse
|
15
|
Kandasamy M, Gileadi U, Rijal P, Tan TK, Lee LN, Chen J, Prota G, Klenerman P, Townsend A, Cerundolo V. Recombinant single-cycle influenza virus with exchangeable pseudotypes allows repeated immunization to augment anti-tumour immunity with immune checkpoint inhibitors. eLife 2023; 12:76414. [PMID: 36626205 PMCID: PMC9831609 DOI: 10.7554/elife.76414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 07/08/2022] [Indexed: 01/11/2023] Open
Abstract
Virus-based tumour vaccines offer many advantages compared to other antigen-delivering systems. They generate concerted innate and adaptive immune response, and robust CD8+ T cell responses. We engineered a non-replicating pseudotyped influenza virus (S-FLU) to deliver the well-known cancer testis antigen, NY-ESO-1 (NY-ESO-1 S-FLU). Intranasal or intramuscular immunization of NY-ESO-1 S-FLU virus in mice elicited a strong NY-ESO-1-specific CD8+ T cell response in lungs and spleen that resulted in the regression of NY-ESO-1-expressing lung tumour and subcutaneous tumour, respectively. Combined administration with anti-PD-1 antibody, NY-ESO-1 S-FLU virus augmented the tumour protection by reducing the tumour metastasis. We propose that the antigen delivery through S-FLU is highly efficient in inducing antigen-specific CD8+ T cell response and protection against tumour development in combination with PD-1 blockade.
Collapse
Affiliation(s)
- Matheswaran Kandasamy
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of OxfordOxfordUnited Kingdom
| | - Uzi Gileadi
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of OxfordOxfordUnited Kingdom
| | - Pramila Rijal
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of OxfordOxfordUnited Kingdom
| | - Tiong Kit Tan
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of OxfordOxfordUnited Kingdom
| | - Lian N Lee
- Nuffield Department of Medicine and Translational Gastroenterology Unit, Peter Medawar Building, University of OxfordOxfordUnited Kingdom
| | - Jili Chen
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of OxfordOxfordUnited Kingdom
| | - Gennaro Prota
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of OxfordOxfordUnited Kingdom
| | - Paul Klenerman
- Nuffield Department of Medicine and Translational Gastroenterology Unit, Peter Medawar Building, University of OxfordOxfordUnited Kingdom
| | - Alain Townsend
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of OxfordOxfordUnited Kingdom
| | - Vincenzo Cerundolo
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of OxfordOxfordUnited Kingdom
| |
Collapse
|
16
|
Jafari M, Kadkhodazadeh M, Shapourabadi MB, Goradel NH, Shokrgozar MA, Arashkia A, Abdoli S, Sharifzadeh Z. Immunovirotherapy: The role of antibody based therapeutics combination with oncolytic viruses. Front Immunol 2022; 13:1012806. [PMID: 36311790 PMCID: PMC9608759 DOI: 10.3389/fimmu.2022.1012806] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/27/2022] [Indexed: 11/13/2022] Open
Abstract
Despite the fact that the new drugs and targeted therapies have been approved for cancer therapy during the past 30 years, the majority of cancer types are still remain challenging to be treated. Due to the tumor heterogeneity, immune system evasion and the complex interaction between the tumor microenvironment and immune cells, the great majority of malignancies need multimodal therapy. Unfortunately, tumors frequently develop treatment resistance, so it is important to have a variety of therapeutic choices available for the treatment of neoplastic diseases. Immunotherapy has lately shown clinical responses in malignancies with unfavorable outcomes. Oncolytic virus (OV) immunotherapy is a cancer treatment strategy that employs naturally occurring or genetically-modified viruses that multiply preferentially within cancer cells. OVs have the ability to not only induce oncolysis but also activate cells of the immune system, which in turn activates innate and adaptive anticancer responses. Despite the fact that OVs were translated into clinical trials, with T-VECs receiving FDA approval for melanoma, their use in fighting cancer faced some challenges, including off-target side effects, immune system clearance, non-specific uptake, and intratumoral spread of OVs in solid tumors. Although various strategies have been used to overcome the challenges, these strategies have not provided promising outcomes in monotherapy with OVs. In this situation, it is increasingly common to use rational combinations of immunotherapies to improve patient benefit. With the development of other aspects of cancer immunotherapy strategies, combinational therapy has been proposed to improve the anti-tumor activities of OVs. In this regard, OVs were combined with other biotherapeutic platforms, including various forms of antibodies, nanobodies, chimeric antigen receptor (CAR) T cells, and dendritic cells, to reduce the side effects of OVs and enhance their efficacy. This article reviews the promising outcomes of OVs in cancer therapy, the challenges OVs face and solutions, and their combination with other biotherapeutic agents.
Collapse
Affiliation(s)
- Mahdie Jafari
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | | | | | - Nasser Hashemi Goradel
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Arash Arashkia
- Department of Molecular Virology, Pasture Institute of Iran, Tehran, Iran
| | - Shahriyar Abdoli
- School of Advanced Medical Technologies, Golestan University of Medical Sciences, Gorgan, Iran
- *Correspondence: Zahra Sharifzadeh, ; Shahriyar Abdoli,
| | - Zahra Sharifzadeh
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
- *Correspondence: Zahra Sharifzadeh, ; Shahriyar Abdoli,
| |
Collapse
|
17
|
Applying nanotechnology to boost cancer immunotherapy by promoting immunogenic cell death. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.10.074] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
18
|
Generation of Human Lung Organoid Cultures from Healthy and Tumor Tissue to Study Infectious Diseases. J Virol 2022; 96:e0009822. [DOI: 10.1128/jvi.00098-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Three-dimensional (3D) human lung organoids reflect the native cell composition of the lung as well as its physiological properties. Human 3D lung organoids offer ideal conditions, such as timely availability in large quantities and high physiological relevance for reassessment and prediction of disease outbreaks of respiratory pathogens and pathogens that use the lung as a primary entry portal.
Collapse
|
19
|
Lei G, Li B, Yang H, Sun F, Li D, Yan J, Wang Y, Li R, Liu H, Zhang S, Li Y, Yang P. Therapeutic Efficacy of an Oncolytic Influenza Virus Carrying an Antibody Against Programmed Cell Death 1 in Hepatocellular Carcinoma. Hum Gene Ther 2022; 33:309-317. [PMID: 35018832 DOI: 10.1089/hum.2021.167] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Oncolytic virus therapy is a promising novel immunotherapy. In this report, we engineered a novel oncolytic influenza virus (IV) carrying an antihuman programmed cell death 1 (PD-1) monoclonal antibody utilizing reverse genetics. A reassortant chimeric IV, named rFlu-huPD1, was synthesized as follows: the heavy chain of the PD-1 antibody was encoded on the PB1 fragment, and the light chain of the PD-1 antibody was encoded on the polymerase acid protein fragment. rFlu-huPD1 antibodies were produced in infected ovalantoic eggs and could replicate to high titers. Moreover, selective cytotoxicity of rFlu-huPD1 was upregulated in multiple hepatocellular carcinoma (HCC) cell lines compared with a control, as determined by a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Furthermore, the activation of T cells in the spleen of tumor-bearing BALB/c mice treated with rFlu-huPD1 was observed, especially cytotoxic CD8+ T cell activation in vivo. In addition, in a patient-derived xenograft liver cancer mouse model, tumor growth was reduced and the overall survival of the mice was increased by intratumoral injections with rFlu-huPD1 compared with wild-type PR8 virus. Taken together, these findings provide evidence for the utility of a combination of oncolytic IVs expressing PD-1 inhibitors for use in HCC virotherapy.
Collapse
Affiliation(s)
- Guanglin Lei
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Baofa Li
- Department of Liver Disease Kaifeng Infectious Disease Hospital, Henan, China
| | - Hao Yang
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Fang Sun
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Donghui Li
- Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jin Yan
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yonggang Wang
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ruisheng Li
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Honghong Liu
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Shaogeng Zhang
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yufeng Li
- First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Penghui Yang
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
20
|
Bu L, Chen B, Xing L, Cai X, Liang S, Zhang L, Wang X, Song W. Generation of a pdmH1N1 2018 Influenza A Reporter Virus Carrying a mCherry Fluorescent Protein in the PA Segment. Front Cell Infect Microbiol 2022; 11:827790. [PMID: 35127568 PMCID: PMC8811159 DOI: 10.3389/fcimb.2021.827790] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 12/31/2021] [Indexed: 11/13/2022] Open
Abstract
Influenza A virus (IAV) is a major human pathogen associated with significant morbidity and mortality worldwide. Through serial passage in mice, we generated a recombinant pdmH1N1 2009 IAV, A/Guangdong/GLW/2018 (GLW/18-MA), which encodes an mCherry gene fused to the C-terminal of a polymerase acidic (PA) segment and demonstrated comparable growth kinetics to the wild-type. Nine mutations were identified in the GLW/18-MA genome: PA (I61M, E351G, and G631S), NP (E292G), HA1 (T164I), HA2 (N117S and P160S), NA (W61R), and NEP (K44R). The recombinant IAV reporter expresses mCherry, a red fluorescent protein, at a high level and maintains its genetic integrity after five generations of serial passages in Madin-Darby Canine Kidney cells (MDCK) cells. Moreover, the imaging is noninvasive and permits the monitoring of infection in living mice. Treatment with oseltamivir or baicalin followed by infection with the reporter IAV led to a decrease in fluorescent protein signal in living mice. This result demonstrates that the IAV reporter virus is a powerful tool to study viral pathogenicity and transmission and to develop and evaluate novel anti-viral drugs, inhibitors, and vaccines in the future.
Collapse
Affiliation(s)
- Ling Bu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Institute of Integration of Traditional and Western Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Boqian Chen
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lei Xing
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Institute of Integration of Traditional and Western Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xuejun Cai
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Institute of Integration of Traditional and Western Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shuhua Liang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Institute of Integration of Traditional and Western Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Liying Zhang
- KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, China
| | - Xinhua Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Institute of Integration of Traditional and Western Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenjun Song
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Institute of Integration of Traditional and Western Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory for Emerging Infectious Diseases, Department of Microbiology, and the Research Center of Infection and Immunology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
21
|
Zhu W, Lv J, Xie X, Tian C, Liu J, Zhou H, Sun C, Li J, Hu Z, Li X. The oncolytic virus VT09X optimizes immune checkpoint therapy in low immunogenic melanoma. Immunol Lett 2021; 241:15-22. [PMID: 34774916 DOI: 10.1016/j.imlet.2021.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 10/26/2021] [Accepted: 11/09/2021] [Indexed: 12/14/2022]
Abstract
Tumors with a low level of pre-existing immune cell infiltration respond poorly to immune checkpoint therapies. Oncolytic viruses optimize immunotherapies by modulating the tumor microenvironment and affecting multiple steps in the cancer-immunity cycle, making them an attractive agent for combination strategies. We engineered an HSV-1-based oncolytic virus and investigated its antitumor effects in combination with the marketed PD-1 antibody Keytruda (pembrolizumab) in hPD-1 knock-in mice bearing non-immunogenic B16-F10 melanoma. Our results showed enhanced CD8+ and CD4+ T cell infiltration, IFN-γ secretion and PD-L1 expression in tumors, subsequently leading to the prolonged overall survival of mice. Systemic changes in lymphocyte cell proportions were also observed in the peripheral blood. In summary, these findings provide evidence that oncolytic viruses can be engineered as a potential platform for combination therapies, especially to treat tumors that are poorly responsive to immune checkpoint therapy.
Collapse
Affiliation(s)
- Wei Zhu
- School of Pharmacy, Yantai University, Yantai 264005, Shandong, China
| | - Jingwen Lv
- School of Pharmacy, Yantai University, Yantai 264005, Shandong, China
| | - Xin Xie
- School of Pharmacy, Yantai University, Yantai 264005, Shandong, China
| | - Chao Tian
- Beijing WellGene Company, Ltd, Beijing 100085, China
| | - Jiajia Liu
- Beijing WellGene Company, Ltd, Beijing 100085, China
| | - Hua Zhou
- Beijing WellGene Company, Ltd, Beijing 100085, China
| | - Chunyang Sun
- Beijing WellGene Company, Ltd, Beijing 100085, China
| | - Jingfeng Li
- School of Pharmacy, Yantai University, Yantai 264005, Shandong, China; Beijing WellGene Company, Ltd, Beijing 100085, China
| | - Zongfeng Hu
- School of Pharmacy, Yantai University, Yantai 264005, Shandong, China
| | - Xiaopeng Li
- School of Pharmacy, Yantai University, Yantai 264005, Shandong, China; Beijing WellGene Company, Ltd, Beijing 100085, China.
| |
Collapse
|
22
|
Kontermann RE, Ungerechts G, Nettelbeck DM. Viro-antibody therapy: engineering oncolytic viruses for genetic delivery of diverse antibody-based biotherapeutics. MAbs 2021; 13:1982447. [PMID: 34747345 PMCID: PMC8583164 DOI: 10.1080/19420862.2021.1982447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Cancer therapeutics approved for clinical application include oncolytic viruses and antibodies, which evolved by nature, but were improved by molecular engineering. Both facilitate outstanding tumor selectivity and pleiotropic activities, but also face challenges, such as tumor heterogeneity and limited tumor penetration. An innovative strategy to address these challenges combines both agents in a single, multitasking therapeutic, i.e., an oncolytic virus engineered to express therapeutic antibodies. Such viro-antibody therapies genetically deliver antibodies to tumors from amplified virus genomes, thereby complementing viral oncolysis with antibody-defined therapeutic action. Here, we review the strategies of viro-antibody therapy that have been pursued exploiting diverse virus platforms, antibody formats, and antibody-mediated modes of action. We provide a comprehensive overview of reported antibody-encoding oncolytic viruses and highlight the achievements of 13 years of viro-antibody research. It has been shown that functional therapeutic antibodies of different formats can be expressed in and released from cancer cells infected with different oncolytic viruses. Virus-encoded antibodies have implemented direct tumor cell killing, anti-angiogenesis, or activation of adaptive immune responses to kill tumor cells, tumor stroma cells or inhibitory immune cells. Importantly, numerous reports have shown therapeutic activity complementary to viral oncolysis for these modalities. Also, challenges for future research have been revealed. Established engineering technologies for both oncolytic viruses and antibodies will enable researchers to address these challenges, facilitating the development of effective viro-antibody therapeutics.
Collapse
Affiliation(s)
- Roland E Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany.,Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| | - Guy Ungerechts
- Clinical Cooperation Unit Virotherapy, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Medical Oncology, National Center for Tumor Diseases (NCT) and University Hospital Heidelberg, Heidelberg, Germany.,Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Dirk M Nettelbeck
- Clinical Cooperation Unit Virotherapy, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
23
|
Wan PKT, Ryan AJ, Seymour LW. Beyond cancer cells: Targeting the tumor microenvironment with gene therapy and armed oncolytic virus. Mol Ther 2021; 29:1668-1682. [PMID: 33845199 PMCID: PMC8116634 DOI: 10.1016/j.ymthe.2021.04.015] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 03/08/2021] [Accepted: 04/06/2021] [Indexed: 01/17/2023] Open
Abstract
Cancer gene therapies are usually designed either to express wild-type copies of tumor suppressor genes or to exploit tumor-associated phenotypic changes to endow selective cytotoxicity. However, these approaches become less relevant to cancers that contain many independent mutations, and the situation is made more complex by our increased understanding of clonal evolution of tumors, meaning that different metastases and even regions of the same tumor mass have distinct mutational and phenotypic profiles. In contrast, the relatively genetically stable tumor microenvironment (TME) therefore provides an appealing therapeutic target, particularly since it plays an essential role in promoting cancer growth, immune tolerance, and acquired resistance to many therapies. Recently, a variety of different TME-targeted gene therapy and armed oncolytic strategies have been explored, with particular success observed in strategies targeting the cancer stroma, reducing tumor vasculature, and repolarizing the immunosuppressive microenvironment. Herein, we review the progress of these TME-targeting approaches and try to highlight those showing the greatest promise.
Collapse
Affiliation(s)
| | - Anderson J Ryan
- Department Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | | |
Collapse
|
24
|
Lei GL, Wang LP, Dong SH, Sun F, Cheng JX, Yang XL, Zhang SG, Wang XL, Wang XX, Yang PH. A recombinant influenza virus with a CTLA4-specific scFv inhibits tumor growth in a mouse model. Cell Biol Int 2021; 45:1202-1210. [PMID: 33501754 DOI: 10.1002/cbin.11559] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 01/04/2021] [Accepted: 01/24/2021] [Indexed: 12/15/2022]
Abstract
Oncolytic viruses (OV) have shown excellent safety and efficacy in preclinical and clinical studies. Influenza A virus (IAV) is considered a promising oncolytic virus. In this report, we generated a recombinant influenza virus expressing an immune checkpoint blockade agent targeting CTLA4. Using reverse genetics, a recombinant influenza virus, termed rFlu-CTLA4, encoding the heavy chain of a CTLA4 antibody on the PB1 segment and the light chain of the CTLA4 antibody on the PA segment was produced. RFlu-CTLA4 could replicate to high titers, and antibodies were produced in the allantoic fluid of infected eggs. Furthermore, the selective cytotoxicity of the virus was higher in various hepatocellular carcinoma cancer cell lines than in the normal cell line L02 in vitro, as indicated by MTS assays. More importantly, in a subcutaneous H22 mouse hepatocarcinoma model, intratumoral injections of rFlu-CTLA4 inhibited the growth of treated tumors and increased the overall survival of mice compared with injections of the PR8 virus. Taken together, these results warrant further exploration of this novel recombinant influenza virus for its potential use as a single or combination agent for cancer immunotherapy.
Collapse
Affiliation(s)
- Guang-Lin Lei
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Li-Peng Wang
- 66400 Military Hospital, Beijing, China.,First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Suo-Hua Dong
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Fang Sun
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jin-Xia Cheng
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiao-Lan Yang
- State Key Laboratory of Pathogens and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Shao-Geng Zhang
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xi-Liang Wang
- State Key Laboratory of Pathogens and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Xin-Xin Wang
- First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Peng-Hui Yang
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
25
|
Masemann D, Meissner R, Schied T, Lichty BD, Rapp UR, Wixler V, Ludwig S. Synergistic anti-tumor efficacy of oncolytic influenza viruses and B7-H3 immune- checkpoint inhibitors against IC-resistant lung cancers. Oncoimmunology 2021; 10:1885778. [PMID: 33643696 PMCID: PMC7894418 DOI: 10.1080/2162402x.2021.1885778] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Non-small cell lung cancers (NSCLCs) establish a highly immunosuppressive tumor microenvironment supporting cancer growth. To interfere with cancer-mediated immunosuppression, selective immune-checkpoint inhibitors (ICIs) have been approved as a standard-of-care treatment for NSCLCs. However, the majority of patients poorly respond to ICI-based immunotherapies. Oncolytic viruses are amongst the many promising immunomodulatory treatments tested as standalone therapy or in combination with ICIs to improve therapeutic outcome. Previously, we demonstrated the oncolytic and immunomodulatory efficacy of low-pathogenic influenza Aviruses (IAVs) against NSCLCs in immunocompetent transgenic mice with alung-specific overexpression of active Raf kinase (Raf-BxB). IAV infection not only resulted in significant primary virus-induced oncolysis, but also caused afunctional reversion of tumor-associated macrophages (TAMs) comprising additional anti-cancer activity. Here we show that NSCLCs as well as TAMs and cytotoxic immune cells overexpress IC molecules of the PD-L2/PD-1 and B7-H3 signaling axes. Thus, we aimed to combine oncolytic IAV-infection with ICIs to exploit the benefits of both anti-cancer approaches. Strikingly, IAV infection combined with the novel B7-H3 ICI led to increased levels of M1-polarized alveolar macrophages and increased lung infiltration by cytotoxic Tlymphocytes, which finally resulted in significantly improved oncolysis of about 80% of existing tumors. In contrast, application of clinically approved α-PD-1 IC antibodies alone or in combination with oncolytic IAV did not provide additional oncolytic or immunomodulatory efficacy. Thus, individualized therapy with synergistically acting oncolytic IAV and B7-H3 ICI might be an innovative future approach to target NSCLCs that are resistant to approved ICIs in patients.
Collapse
Affiliation(s)
- Dörthe Masemann
- Cells in Motion" Interfaculty Center (Cimic), University of Muenster, Muenster, Germany.,Center for Molecular Biology of Inflammation, Institute of Virology, University of Muenster, Muenster, Germany
| | - Ramona Meissner
- Cells in Motion" Interfaculty Center (Cimic), University of Muenster, Muenster, Germany.,Center for Molecular Biology of Inflammation, Institute of Virology, University of Muenster, Muenster, Germany
| | - Tanja Schied
- Center for Molecular Biology of Inflammation, Institute of Virology, University of Muenster, Muenster, Germany
| | - Brian D Lichty
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Ulf R Rapp
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Viktor Wixler
- Cells in Motion" Interfaculty Center (Cimic), University of Muenster, Muenster, Germany.,Center for Molecular Biology of Inflammation, Institute of Virology, University of Muenster, Muenster, Germany
| | - Stephan Ludwig
- Cells in Motion" Interfaculty Center (Cimic), University of Muenster, Muenster, Germany.,Center for Molecular Biology of Inflammation, Institute of Virology, University of Muenster, Muenster, Germany
| |
Collapse
|
26
|
Development and application of reverse genetic technology for the influenza virus. Virus Genes 2021; 57:151-163. [PMID: 33528730 PMCID: PMC7851324 DOI: 10.1007/s11262-020-01822-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/28/2020] [Indexed: 12/28/2022]
Abstract
Influenza virus is a common virus in people's daily lives, and it has certain infectivity in humans and animals. Influenza viruses have the characteristics of a high mutation rate and wide distribution. Reverse genetic technology is primarily used to modify viruses at the DNA level through targeted modification of the virus cDNA. Genetically modified influenza viruses have a unique advantage when researching the transmission and pathogenicity of influenza. With the continuous development of oncolytic viruses in recent years, studies have found that influenza viruses also have certain oncolytic activity. Influenza viruses can specifically recognize tumor cells; activate cytotoxic T cells, NK cells, dendritic cells, etc.; and stimulate the body to produce an immune response, thereby killing tumor cells. This article will review the development and application of influenza virus reverse genetic technology.
Collapse
|
27
|
tenOever BR. Synthetic Virology: Building Viruses to Better Understand Them. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a038703. [PMID: 31871242 DOI: 10.1101/cshperspect.a038703] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Generally comprised of less than a dozen components, RNA viruses can be viewed as well-designed genetic circuits optimized to replicate and spread within a given host. Understanding the molecular design that enables this activity not only allows one to disrupt these circuits to study their biology, but it provides a reprogramming framework to achieve novel outputs. Recent advances have enabled a "learning by building" approach to better understand virus biology and create valuable tools. Below is a summary of how modifying the preexisting genetic framework of influenza A virus has been used to track viral movement, understand virus replication, and identify host factors that engage this viral circuitry.
Collapse
Affiliation(s)
- Benjamin R tenOever
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| |
Collapse
|
28
|
Hu PY, Fan XM, Zhang YN, Wang SB, Wan WJ, Pan HY, Mou XZ. The limiting factors of oncolytic virus immunotherapy and the approaches to overcome them. Appl Microbiol Biotechnol 2020; 104:8231-8242. [PMID: 32816087 DOI: 10.1007/s00253-020-10802-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 06/22/2020] [Accepted: 07/30/2020] [Indexed: 02/07/2023]
Abstract
Oncolytic virus (OV) immunotherapy is characterized by viruses which specifically target cancer cells and cause their cytolysis. They provide a unique and promising new tool for the eradication of cancer as they interact with and affect the tumor microenvironment (TME), vasculature, and immune system. Advancements of genetic engineering have allowed for these viruses to be armed in such a way to have enhanced targeting, strong immunomodulation properties, and an ability to modify the TME. However, there are still major limitations in their use, mostly due to difficulties in delivering the viral particles to the tumors and in ensuring that the immunomodulatory properties are able to stimulate the host immune response to mount a complete response. Using novel delivery systems and using OVs as a complementary therapy in a combinatorial treatment have shown some significant successes. In this review, we discuss the major issues and difficulties in using OVs as anti-tumor agents and some of the strategies put in place so far to overcome these limitations. KEY POINTS: • Oncolytic viruses (OVs) infect cancer cells and cause their cytolysis. • The major limitations in using OVs as anti-tumor therapy were discussed. • The potential strategies to overcome these limitations were summarized.
Collapse
Affiliation(s)
- Pei-Yang Hu
- Department of Traumatology, Tiantai People's Hospital of Zhejiang Province (Tiantai Branch of Zhejiang People's Hospital), Taizhou, 317200, China
| | - Xiao-Ming Fan
- Department of Ultrasound, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, 310014, China
| | - You-Ni Zhang
- Department of Traumatology, Tiantai People's Hospital of Zhejiang Province (Tiantai Branch of Zhejiang People's Hospital), Taizhou, 317200, China.,Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, 310014, China.,Clinical Research Institute, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, 310014, China
| | - Shi-Bing Wang
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, 310014, China.,Clinical Research Institute, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, 310014, China
| | - Wei-Jie Wan
- Shandong Xiandai University, Jinan, 250104, China
| | - Hong-Ying Pan
- Department of Infectious Diseases, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, 310014, China.
| | - Xiao-Zhou Mou
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, 310014, China. .,Clinical Research Institute, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, 310014, China.
| |
Collapse
|
29
|
Kabiljo J, Laengle J, Bergmann M. From threat to cure: understanding of virus-induced cell death leads to highly immunogenic oncolytic influenza viruses. Cell Death Discov 2020; 6:48. [PMID: 32542113 PMCID: PMC7288254 DOI: 10.1038/s41420-020-0284-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 02/18/2020] [Accepted: 02/20/2020] [Indexed: 01/08/2023] Open
Abstract
Oncolytic viruses constitute an emerging strategy in immunomodulatory cancer treatment. The first oncolytic virus, Talimogene laherparepvec (T-VEC), based on herpes simplex virus 1 (HSV-1), was approved by the Food and Drug Administration (FDA) and European Medicines Agency (EMA) in 2015. The field of oncolytic virotherapy is still in its beginnings, since many promising viruses remain only superficially explored. Influenza A virus causes a highly immunogenic acute infection but never leads to a chronic disease. While oncolytic influenza A viruses are in preclinical development, they have not made the transition into clinical practice yet. Recent insights into different types of cell death caused by influenza A virus infection illuminate novel possibilities of enhancing its therapeutic effect. Genetic engineering and experience in influenza A virus vaccine development allow safe application of the virus in patients. In this review we give a summary of efforts undertaken to develop oncolytic influenza A viruses. We discuss strategies for targeting viral replication to cancerous lesions and arming them with immunogenic transgenes. We furthermore describe which modes of cell death are induced by influenza A virus infection and how these insights may be utilized to optimize influenza A virus-based oncolytic virus design.
Collapse
Affiliation(s)
- Julijan Kabiljo
- Division of General Surgery, Department of Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Johannes Laengle
- Division of General Surgery, Department of Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Michael Bergmann
- Division of General Surgery, Department of Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| |
Collapse
|
30
|
Page A, Fusil F, Cosset FL. Towards Physiologically and Tightly Regulated Vectored Antibody Therapies. Cancers (Basel) 2020; 12:E962. [PMID: 32295072 PMCID: PMC7226531 DOI: 10.3390/cancers12040962] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/09/2020] [Accepted: 04/10/2020] [Indexed: 12/20/2022] Open
Abstract
Cancers represent highly significant health issues and the options for their treatment are often not efficient to cure the disease. Immunotherapy strategies have been developed to modulate the patient's immune system in order to eradicate cancerous cells. For instance, passive immunization consists in the administration at high doses of exogenously produced monoclonal antibodies directed either against tumor antigen or against immune checkpoint inhibitors. Its main advantage is that it provides immediate immunity, though during a relatively short period, which consequently requires frequent injections. To circumvent this limitation, several approaches, reviewed here, have emerged to induce in vivo antibody secretion at physiological doses. Gene delivery vectors, such as adenoviral vectors or adeno-associated vectors, have been designed to induce antibody secretion in vivo after in situ cell modification, and have driven significant improvements in several cancer models. However, anti-idiotypic antibodies and escape mutants have been detected, probably because of both the continuous expression of antibodies and their expression by unspecialized cell types. To overcome these hurdles, adoptive transfer of genetically modified B cells that secrete antibodies either constitutively or in a regulated manner have been developed by ex vivo transgene insertion with viral vectors. Recently, with the emergence of gene editing technologies, the endogenous B cell receptor loci of B cells have been modified with the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated endonuclease (Cas-9) system to change their specificity in order to target a given antigen. The expression of the modified BCR gene hence follows the endogenous regulation mechanisms, which may prevent or at least reduce side effects. Although these approaches seem promising for cancer treatments, major questions, such as the persistence and the re-activation potential of these engineered cells, remain to be addressed in clinically relevant animal models before translation to humans.
Collapse
Affiliation(s)
| | | | - François-Loïc Cosset
- CIRICentre International de Recherche en Infectiologie, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, 46 allée d’Italie, F-69007 Lyon, France; (A.P.); (F.F.)
| |
Collapse
|
31
|
PD-1 IC Inhibition Synergistically Improves Influenza A Virus-Mediated Oncolysis of Metastatic Pulmonary Melanoma. MOLECULAR THERAPY-ONCOLYTICS 2020; 17:190-204. [PMID: 32346609 PMCID: PMC7178321 DOI: 10.1016/j.omto.2020.03.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 03/30/2020] [Indexed: 01/02/2023]
Abstract
Recently, we showed that infection of primary lung tumor-bearing mice with oncolytic influenza A viruses (IAVs) led to strong virus-induced tumor cell lysis but also to restoration of immune competence of innate immune cells. Murine B16-F10 melanoma cells are known for their high lung tropism and progressive growth. As these cells are also highly permissive for IAVs, we analyzed their oncolytic and immunomodulatory efficiency against pulmonary B16-F10 lung metastases in vivo. IAV infection abrogated the melanoma-mediated immune suppression in the lung and induced a more than 50% cancer cell lysis. The oncolytic effect reached maximal efficacy 3 days post-infection, but it was not sustained over time. In order to maintain the virus-induced anti-tumor effect, mice with melanoma-derived lung cancers were treated in addition to influenza virus infection with an immune checkpoint inhibitor against programmed death-1 receptor (PD-1). The combined IAV and immune checkpoint inhibition (ICI) therapy resulted in a sustained anti-tumor efficacy, keeping the lung melanoma mass at day 12 of IAV infection still reduced by 50% over the control mice. In conclusion, ICI treatment strongly enhanced the oncolytic effect of influenza virus infection, suggesting that combined treatment is a promising approach against metastatic pulmonary melanoma.
Collapse
|
32
|
Jacobs L, De Smidt E, Geukens N, Declerck P, Hollevoet K. DNA-Based Delivery of Checkpoint Inhibitors in Muscle and Tumor Enables Long-Term Responses with Distinct Exposure. Mol Ther 2020; 28:1068-1077. [PMID: 32101701 DOI: 10.1016/j.ymthe.2020.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 02/08/2020] [Indexed: 12/17/2022] Open
Abstract
Checkpoint-inhibiting antibodies elicit impressive clinical responses, but still face several issues. The current study evaluated whether DNA-based delivery can broaden the application of checkpoint inhibitors, specifically by pursuing cost-efficient in vivo production, facilitating combination therapies, and exploring administration routes that lower immune-related toxicity risks. We therefore optimized plasmid-encoded anti-CTLA-4 and anti-PD-1 antibodies, and studied their pharmacokinetics and pharmacodynamics when delivered alone and in combination via intramuscular or intratumoral electroporation in mice. Intramuscular electrotransfer of these DNA-based antibodies induced complete regressions in a subcutaneous MC38 tumor model, with plasma concentrations up to 4 and 14 μg/mL for anti-CTLA-4 and anti-PD-1 antibodies, respectively, and antibody detection for at least 6 months. Intratumoral antibody gene electrotransfer gave similar anti-tumor responses as the intramuscular approach. Antibody plasma levels, however, were up to 70-fold lower and substantially more transient, potentially improving biosafety of the expressed checkpoint inhibitors. Intratumoral delivery also generated a systemic anti-tumor response, illustrated by moderate abscopal effects and prolonged protection of cured mice against a tumor rechallenge. In conclusion, intramuscular and intratumoral DNA-based delivery of checkpoint inhibitors both enabled long-term anti-tumor responses despite distinct systemic antibody exposure, highlighting the potential of the tumor as delivery site for DNA-based therapeutics.
Collapse
Affiliation(s)
- Liesl Jacobs
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven - University of Leuven, Leuven, Belgium
| | - Elien De Smidt
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven - University of Leuven, Leuven, Belgium; PharmAbs - The KU Leuven Antibody Center, KU Leuven - University of Leuven, Leuven, Belgium
| | - Nick Geukens
- PharmAbs - The KU Leuven Antibody Center, KU Leuven - University of Leuven, Leuven, Belgium
| | - Paul Declerck
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven - University of Leuven, Leuven, Belgium; PharmAbs - The KU Leuven Antibody Center, KU Leuven - University of Leuven, Leuven, Belgium.
| | - Kevin Hollevoet
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven - University of Leuven, Leuven, Belgium.
| |
Collapse
|
33
|
A Recombinant Influenza A/H1N1 Carrying A Short Immunogenic Peptide of MERS-CoV as Bivalent Vaccine in BALB/c Mice. Pathogens 2019; 8:pathogens8040281. [PMID: 31810359 PMCID: PMC6963271 DOI: 10.3390/pathogens8040281] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/21/2019] [Accepted: 11/30/2019] [Indexed: 01/26/2023] Open
Abstract
Middle East Respiratory Syndrome Coronavirus (MERS-CoV) became a global human health threat since its first documentation in humans in 2012. An efficient vaccine for the prophylaxis of humans in hotspots of the infection (e.g., Saudi Arabia) is necessary but no commercial vaccines are yet approved. In this study, a chimeric DNA construct was designed to encode an influenza A/H1N1 NA protein which is flanking immunogenic amino acids (aa) 736–761 of MERS-CoV spike protein. Using the generated chimeric construct, a novel recombinant vaccine strain against pandemic influenza A virus (H1N1pdm09) and MERS-CoV was generated (chimeric bivalent 5 + 3). The chimeric bivalent 5 + 3 vaccine strain comprises a recombinant PR8-based vaccine, expressing the PB1, HA, and chimeric NA of pandemic 2009 H1N1. Interestingly, an increase in replication efficiency of the generated vaccine strain was observed when compared to the PR8-based 5 + 3 H1N1pdm09 vaccine strain that lacks the MERS-CoV spike peptide insert. In BALB/c mice, the inactivated chimeric bivalent vaccine induced potent and specific neutralizing antibodies against MERS-CoV and H1N1pdm09. This novel approach succeeded in developing a recombinant influenza virus with potential use as a bivalent vaccine against H1N1pdm09 and MERS-CoV. This approach provides a basis for the future development of chimeric influenza-based vaccines against MERS-CoV and other viruses.
Collapse
|
34
|
Kim SY, Kim S, Kim JE, Lee SN, Shin IW, Shin HS, Jin SM, Noh YW, Kang YJ, Kim YS, Kang TH, Park YM, Lim YT. Lyophilizable and Multifaceted Toll-like Receptor 7/8 Agonist-Loaded Nanoemulsion for the Reprogramming of Tumor Microenvironments and Enhanced Cancer Immunotherapy. ACS NANO 2019; 13:12671-12686. [PMID: 31589013 DOI: 10.1021/acsnano.9b04207] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The low therapeutic efficacy of current cancer immunotherapy is related to nonimmunogenic and immunosuppressive tumor microenvironments (TMEs). To overcome these limitations, both the immune priming of antitumoral lymphocytes and the reprogramming of immunosuppressive factors in TMEs are essential. Here, we suggest a nanoemulsion (NE)-based immunotherapeutic platform that can not only modulate tumor-induced suppression but also induce an effective cell-mediated immune response for T cell proliferation. Multifunctional NEs can be fabricated by integrating the efficacy of NEs as delivery systems and the multifaceted immunomodulation characteristics (i.e., immunostimulation and reprogramming of immunosuppression) of small molecule-based Toll-like receptor 7/8 agonists. Local in situ vaccination of melanoma and cervical tumor models with tumor antigens (protein and peptide) adjuvanted with NE loaded with TLR7/8 agonists [NE (TLR7/8a)] induced the recruitment and activation of innate immune cells, infiltration of lymphocytes, and polarization of tumor-associated M2 macrophages, which resulted in inhibition of tumor growth and prolonged survival in both primary and rechallenged tumor models. Antibody-depletion experiments also suggested that macrophages, type I IFN (IFN-α and IFN-β), CD8+ T cells, and NK1.1+ cells contributed to the antitumor effect of NE (TLR7/8a). The combination of antitumoral lymphocytes and reprogramming of immunosuppressive TMEs induced by NE (TLR7/8a) treatment evoked a synergistic antitumor immune response with immune checkpoint blockade therapy (anti-PD-1 and anti-PD-L1).
Collapse
Affiliation(s)
- Sun-Young Kim
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, School of Chemical Engineering , Sungkyunkwan University (SKKU) , 2066 Seobu-ro , Jangan-gu, Suwon , Gyeonggi-do 16419 , Republic of Korea
| | - Sohyun Kim
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, School of Chemical Engineering , Sungkyunkwan University (SKKU) , 2066 Seobu-ro , Jangan-gu, Suwon , Gyeonggi-do 16419 , Republic of Korea
| | - Jung-Eun Kim
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, School of Chemical Engineering , Sungkyunkwan University (SKKU) , 2066 Seobu-ro , Jangan-gu, Suwon , Gyeonggi-do 16419 , Republic of Korea
| | - Sang Nam Lee
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, School of Chemical Engineering , Sungkyunkwan University (SKKU) , 2066 Seobu-ro , Jangan-gu, Suwon , Gyeonggi-do 16419 , Republic of Korea
| | - Il Woo Shin
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, School of Chemical Engineering , Sungkyunkwan University (SKKU) , 2066 Seobu-ro , Jangan-gu, Suwon , Gyeonggi-do 16419 , Republic of Korea
| | - Hong Sik Shin
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, School of Chemical Engineering , Sungkyunkwan University (SKKU) , 2066 Seobu-ro , Jangan-gu, Suwon , Gyeonggi-do 16419 , Republic of Korea
| | - Seung Mo Jin
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, School of Chemical Engineering , Sungkyunkwan University (SKKU) , 2066 Seobu-ro , Jangan-gu, Suwon , Gyeonggi-do 16419 , Republic of Korea
| | - Young-Woock Noh
- New Drug Development Center , Osong Medical Innovation Foundation , 123 Osongsaengmyeong-ro , Cheongju , Chungcheongbuk-do 28160 , Republic of Korea
| | - Young Ju Kang
- New Drug Development Center , Osong Medical Innovation Foundation , 123 Osongsaengmyeong-ro , Cheongju , Chungcheongbuk-do 28160 , Republic of Korea
| | - Young Seob Kim
- Department of Immunology, School of Medicine , Konkuk University , 268 Chungwondae-ro , Chungju , Chungcheongbuk-do 27478 , Republic of Korea
| | - Tae Heung Kang
- Department of Immunology, School of Medicine , Konkuk University , 268 Chungwondae-ro , Chungju , Chungcheongbuk-do 27478 , Republic of Korea
| | - Yeong-Min Park
- Department of Immunology, School of Medicine , Konkuk University , 268 Chungwondae-ro , Chungju , Chungcheongbuk-do 27478 , Republic of Korea
| | - Yong Taik Lim
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, School of Chemical Engineering , Sungkyunkwan University (SKKU) , 2066 Seobu-ro , Jangan-gu, Suwon , Gyeonggi-do 16419 , Republic of Korea
| |
Collapse
|
35
|
Aleynick M, Svensson-Arvelund J, Flowers CR, Marabelle A, Brody JD. Pathogen Molecular Pattern Receptor Agonists: Treating Cancer by Mimicking Infection. Clin Cancer Res 2019; 25:6283-6294. [PMID: 31123052 PMCID: PMC11734666 DOI: 10.1158/1078-0432.ccr-18-1800] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/08/2019] [Accepted: 05/16/2019] [Indexed: 11/16/2022]
Abstract
Immunotherapies such as checkpoint blockade have achieved durable benefits for patients with advanced stage cancer and have changed treatment paradigms. However, these therapies rely on a patient's own a priori primed tumor-specific T cells, limiting their efficacy to a subset of patients. Because checkpoint blockade is most effective in patients with inflamed or "hot" tumors, a priority in the field is learning how to "turn cold tumors hot." Inflammation is generally initiated by innate immune cells, which receive signals through pattern recognition receptors (PRR)-a diverse family of receptors that sense conserved molecular patterns on pathogens, alarming the immune system of an invading microbe. Their immunostimulatory properties can reprogram the immune suppressive tumor microenvironment and activate antigen-presenting cells to present tumors antigens, driving de novo tumor-specific T-cell responses. These features, among others, make PRR-targeting therapies an attractive strategy in immuno-oncology. Here, we discuss mechanisms of PRR activation, highlighting ongoing clinical trials and recent preclinical advances focused on therapeutically targeting PRRs to treat cancer.
Collapse
Affiliation(s)
- Mark Aleynick
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | - Christopher R Flowers
- Winship Cancer Institute of Emory University, Emory University School of Medicine, Atlanta, Georgia
| | | | - Joshua D Brody
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
- Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
36
|
Vijayakumar G, Palese P, Goff PH. Oncolytic Newcastle disease virus expressing a checkpoint inhibitor as a radioenhancing agent for murine melanoma. EBioMedicine 2019; 49:96-105. [PMID: 31676387 PMCID: PMC6945240 DOI: 10.1016/j.ebiom.2019.10.032] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/09/2019] [Accepted: 10/16/2019] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Monoclonal antibodies (mAbs) targeting negative regulators, or checkpoint molecules (e.g. PD1/PD-L1 & CTLA4), of anti-tumoural T cells have demonstrated clinical efficacy in treating several neoplastic diseases. While many patients enjoy remarkable responses to checkpoint inhibitors, a majority show adverse effects. Understanding how checkpoint inhibitors may augment established chemotherapy or radiotherapy regimens or other immunotherapies like oncolytic viruses may lead to better clinical outcomes measured by improved efficacy with reduced toxicity. Here, we assess how Newcastle disease virus (NDV), an oncolytic virus in clinical testing, may interact with radiotherapy to enhance checkpoint inhibitor blockade. METHODS An immunocompetent B16-F10 murine melanoma model, generally considered to be a poorly immunogenic or "cold" tumour, was utilised to query whether combining localised radiotherapy with NDV may be more effective than either therapy alone in controlling tumours in mice treated with anti-PD1 or anti-CTLA4 monoclonal antibodies. We also investigated whether localised administration of a checkpoint inhibitor through an intratumoural injection of NDV that expresses anti-CTLA4 single-chain variable fragment (scFv) is comparable to systemic administration of anti-CTLA4 when combined with radiation in mediating its anti-tumour efficacy. Response rates were characterised by measuring tumour size over time, observation of complete tumour regression, and overall survival. FINDINGS Our results show that combining NDV plus radiotherapy with checkpoint inhibitors (PD1 or CTLA4 targeted mAbs) results in significantly better complete tumour regression rates with an abscopal effect in a murine model of melanoma than either single therapy combined with checkpoint inhibitors. Finally, we also show that localised administration of a recombinant NDV expressing anti-CTLA4 plus radiation is comparable to systemic anti-CTLA4 plus radiation in mediating its anti-tumour effect as assayed by survival benefit. INTERPRETATION Our results show that oncolytic NDV plus radiotherapy work together with checkpoint inhibitors to enhance tumour clearance of murine melanoma. NDV is an effective radiotherapy dose-sparing and immunotherapeutic agent capable of transgenic, in vivo expression of an anti-CTLA4 targeted scFv antibody with the potential to spare systemic exposure. FUNDING The National Institutes of Health grant HHSN272201400008C supported the work. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
Collapse
Affiliation(s)
- Gayathri Vijayakumar
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Peter Palese
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Peter H Goff
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Radiation Oncology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
37
|
Harrington K, Freeman DJ, Kelly B, Harper J, Soria JC. Optimizing oncolytic virotherapy in cancer treatment. Nat Rev Drug Discov 2019; 18:689-706. [PMID: 31292532 DOI: 10.1038/s41573-019-0029-0] [Citation(s) in RCA: 337] [Impact Index Per Article: 56.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2019] [Indexed: 02/07/2023]
Abstract
In the wake of the success of modern immunotherapy, oncolytic viruses (OVs) are currently seen as a potential therapeutic option for patients with cancer who do not respond or fail to achieve durable responses following treatment with immune checkpoint inhibitors. OVs offer a multifaceted therapeutic platform because they preferentially replicate in tumour cells, can be engineered to express transgenes that augment their cytotoxic and immunostimulatory activities, and modulate the tumour microenvironment to optimize immune-mediated tumour eradication, both at locoregional and systemic sites of disease. Lysis of tumour cells releases tumour-specific antigens that trigger both the innate and adaptive immune systems. OVs also represent attractive combination partners with other systemically delivered agents by virtue of their highly favourable safety profiles. Rational combinations of OVs with different immune modifiers and/or antitumour agents, based on mechanisms of tumour resistance to immune-mediated attack, may benefit the large, currently underserved, population of patients who respond poorly to immune checkpoint inhibition.
Collapse
Affiliation(s)
- Kevin Harrington
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK.
| | | | - Beth Kelly
- Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | | | - Jean-Charles Soria
- Oncology R&D, AstraZeneca, Gaithersburg, MD, USA.,Department of Medicine and Medical Oncology, Université Paris-Sud, Orsay, France
| |
Collapse
|
38
|
Behzadi MA, Stein KR, Bermúdez-González MC, Simon V, Nachbagauer R, Tortorella D. An Influenza Virus Hemagglutinin-Based Vaccine Platform Enables the Generation of Epitope Specific Human Cytomegalovirus Antibodies. Vaccines (Basel) 2019; 7:vaccines7020051. [PMID: 31207917 PMCID: PMC6630953 DOI: 10.3390/vaccines7020051] [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: 05/06/2019] [Revised: 06/06/2019] [Accepted: 06/10/2019] [Indexed: 12/03/2022] Open
Abstract
Human cytomegalovirus (CMV) is a highly prevalent pathogen with ~60%–90% seropositivity in adults. CMV can contribute to organ rejection in transplant recipients and is a major cause of birth defects in newborns. Currently, there are no approved vaccines against CMV. The epitope of a CMV neutralizing monoclonal antibody against a conserved region of the envelope protein gH provided the basis for a new CMV vaccine design. We exploited the influenza A virus as a vaccine platform due to the highly immunogenic head domain of its hemagglutinin envelope protein. Influenza A variants were engineered by reverse genetics to express the epitope of an anti-CMV gH neutralizing antibody that recognizes native gH into the hemagglutinin antigenic Sa site. We determined that the recombinant influenza variants expressing 7, 10, or 13 residues of the anti-gH neutralizing antibody epitope were recognized and neutralized by the anti-gH antibody 10C10. Mice vaccinated with the influenza/CMV chimeric viruses induced CMV-specific antibodies that recognized the native gH protein and inhibited virus infection. In fact, the influenza variants expressing 7–13 gH residues neutralized a CMV infection at ~60% following two immunizations with variants expressing the 13 residue gH peptide produced the highest levels of neutralization. Collectively, our study demonstrates that a variant influenza virus inserted with a gH peptide can generate a humoral response that limits a CMV infection.
Collapse
Affiliation(s)
- Mohammad Amin Behzadi
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Kathryn R Stein
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Maria Carolina Bermúdez-González
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
- The Global Health Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Viviana Simon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
- The Global Health Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
- Division of Infectious Disease, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Raffael Nachbagauer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Domenico Tortorella
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
39
|
Crupi MJF, Bell JC, Singaravelu R. Concise Review: Targeting Cancer Stem Cells and Their Supporting Niche Using Oncolytic Viruses. Stem Cells 2019; 37:716-723. [PMID: 30875126 DOI: 10.1002/stem.3004] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 02/08/2019] [Accepted: 03/02/2019] [Indexed: 12/22/2022]
Abstract
Cancer stem cells (CSCs) have the capacity to self-renew and differentiate to give rise to heterogenous cancer cell lineages in solid tumors. These CSC populations are associated with metastasis, tumor relapse, and resistance to conventional anticancer therapies. Here, we focus on the use of oncolytic viruses (OVs) to target CSCs as well as the OV-driven interferon production in the tumor microenvironment (TME) that can repress CSC properties. We explore the ability of OVs to deliver combinations of immune-modulating therapeutic transgenes, such as immune checkpoint inhibitor antibodies. In particular, we highlight the advantages of virally encoded bi-specific T cell engagers (BiTEs) to not only target cell-surface markers on CSCs, but also tumor-associated antigens on contributing components of the surrounding TME and other cancer cells. We also highlight the crucial role of combination anticancer treatments, evidenced by synergy of OV-delivered BiTEs and chimeric-antigen receptor T cell therapy. Stem Cells 2019;37:716-723.
Collapse
Affiliation(s)
- Mathieu J F Crupi
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - John C Bell
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Ragunath Singaravelu
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
40
|
Gerlach T, Elbahesh H, Saletti G, Rimmelzwaan GF. Recombinant influenza A viruses as vaccine vectors. Expert Rev Vaccines 2019; 18:379-392. [PMID: 30777467 DOI: 10.1080/14760584.2019.1582338] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Various viruses, including poxviruses, adenoviruses and vesicular stomatitis virus, have been considered as vaccine vectors for the delivery of antigens of interest in the development of vaccines against newly emerging pathogens. AREAS COVERED Here, we review results that have been obtained with influenza A viruses (IAV) as vaccine vectors. With the advent of reverse genetics technology, IAV-based recombinant vaccine candidates have been constructed that induce protective immunity to a variety of different pathogens of interest, including West Nile virus, Plasmodium falciparum and respiratory syncytial virus. The various cloning strategies to produce effective and attenuated, safe to use IAV-based viral vectors are discussed. EXPERT COMMENTARY It was concluded that IAV-based vector system has several advantages and holds promise for further development.
Collapse
Affiliation(s)
- Thomas Gerlach
- a Research Center for Emerging Infections and Zoonoses (RIZ) , University of Veterinary Medicine Hannover (TiHo) , Hannover , Germany
| | - Husni Elbahesh
- a Research Center for Emerging Infections and Zoonoses (RIZ) , University of Veterinary Medicine Hannover (TiHo) , Hannover , Germany
| | - Giulietta Saletti
- a Research Center for Emerging Infections and Zoonoses (RIZ) , University of Veterinary Medicine Hannover (TiHo) , Hannover , Germany
| | - Guus F Rimmelzwaan
- a Research Center for Emerging Infections and Zoonoses (RIZ) , University of Veterinary Medicine Hannover (TiHo) , Hannover , Germany
| |
Collapse
|
41
|
Penghui Y, Fang S, Ruilan W, Guanglin L, Hongjing G, Yueqiang D, Zhongpeng Z, Xiaolan Y, Zhaohai W, Shaogeng Z, Xiliang W. Oncolytic Activity of a Novel Influenza A Virus Carrying Granulocyte-Macrophage Colony-Stimulating Factor in Hepatocellular Carcinoma. Hum Gene Ther 2018; 30:330-338. [PMID: 30205709 DOI: 10.1089/hum.2018.095] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Oncolytic virotherapy is a promising strategy for the treatment of cancer. Influenza A virus has shown potential as an oncolytic agent. In this study, a recombinant PR8 influenza viral vector, called delNS1-GM-CSF, was generated with a partial deletion in NS and the granulocyte-macrophage colony-stimulating factor (GM-CSF) coding sequence inserted into the influenza nonstructural protein 1 gene. The morphological characteristics of delNS1-GM-CSF were examined. The delNS1-GM-CSF virus replicated well in various cell lines, including MDCK, A549, SMCC7721, and HepG2 cells. Moreover, selective cytotoxicity of the virus was observed in various hepatocellular carcinoma (HCC) cell lines, while no effect was demonstrated in the normal liver cell line LO2, as indicated by 3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyltetrazolium bromide and crystal violet assays. Importantly, using a model based on the growth of HepG2 cells as a xenograft in nude mice, it was found that a reassortant delNS1-GM-CSF virus inhibited tumor growth significantly following intratumoral injection in a dose-dependent manner. Ex vivo results showed that the tumor inhibition efficacy of delNS1-GM-CSF was observed in HCC clinical samples. Taken together, these results are the first to demonstrate that influenza A viruses may have potential as oncolytic virotherapeutic agents against HCC.
Collapse
Affiliation(s)
- Yang Penghui
- 1 Department of Hepatobiliary, Beijing 302 Hospital, Beijing, P.R. China.,2 State Key Laboratory of Pathogens and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, P.R. China
| | - Sun Fang
- 1 Department of Hepatobiliary, Beijing 302 Hospital, Beijing, P.R. China
| | - Wang Ruilan
- 1 Department of Hepatobiliary, Beijing 302 Hospital, Beijing, P.R. China
| | - Lei Guanglin
- 1 Department of Hepatobiliary, Beijing 302 Hospital, Beijing, P.R. China
| | - Gu Hongjing
- 2 State Key Laboratory of Pathogens and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, P.R. China
| | - Duan Yueqiang
- 2 State Key Laboratory of Pathogens and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, P.R. China
| | - Zhao Zhongpeng
- 2 State Key Laboratory of Pathogens and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, P.R. China
| | - Yang Xiaolan
- 2 State Key Laboratory of Pathogens and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, P.R. China
| | - Wang Zhaohai
- 1 Department of Hepatobiliary, Beijing 302 Hospital, Beijing, P.R. China
| | - Zhang Shaogeng
- 1 Department of Hepatobiliary, Beijing 302 Hospital, Beijing, P.R. China
| | - Wang Xiliang
- 2 State Key Laboratory of Pathogens and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, P.R. China
| |
Collapse
|
42
|
Pol JG, Lévesque S, Workenhe ST, Gujar S, Le Boeuf F, Clements DR, Fahrner JE, Fend L, Bell JC, Mossman KL, Fucikova J, Spisek R, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Oncolytic viro-immunotherapy of hematologic and solid tumors. Oncoimmunology 2018; 7:e1503032. [PMID: 30524901 PMCID: PMC6279343 DOI: 10.1080/2162402x.2018.1503032] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 07/15/2018] [Indexed: 02/08/2023] Open
Abstract
Oncolytic viruses selectively target and kill cancer cells in an immunogenic fashion, thus supporting the establishment of therapeutically relevant tumor-specific immune responses. In 2015, the US Food and Drug Administration (FDA) approved the oncolytic herpes simplex virus T-VEC for use in advanced melanoma patients. Since then, a plethora of trials has been initiated to assess the safety and efficacy of multiple oncolytic viruses in patients affected with various malignancies. Here, we summarize recent preclinical and clinical progress in the field of oncolytic virotherapy.
Collapse
Affiliation(s)
- Jonathan G. Pol
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
| | - Sarah Lévesque
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
| | - Samuel T. Workenhe
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Shashi Gujar
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
- Department of Microbiology and Immunology, Dalhousie University, NS, Canada
- Department of Biology, Dalhousie University, NS, Canada
- Centre for Innovative and Collaborative Health Sciences Research, Quality and System Performance, IWK Health Centre, Halifax, NS, Canada
| | - Fabrice Le Boeuf
- Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | | | - Jean-Eudes Fahrner
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, Villejuif, France
- Transgene S.A., Illkirch-Graffenstaden, France
| | | | - John C. Bell
- Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Karen L. Mossman
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Jitka Fucikova
- Sotio a.c., Prague, Czech Republic
- Department of Immunology, 2nd Faculty of Medicine, University Hospital Motol, Charles University, Prague, Czech Republic
| | - Radek Spisek
- Sotio a.c., Prague, Czech Republic
- Department of Immunology, 2nd Faculty of Medicine, University Hospital Motol, Charles University, Prague, Czech Republic
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, Villejuif, France
| | - Guido Kroemer
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, Paris, France
- Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
43
|
Leoni V, Vannini A, Gatta V, Rambaldi J, Sanapo M, Barboni C, Zaghini A, Nanni P, Lollini PL, Casiraghi C, Campadelli-Fiume G. A fully-virulent retargeted oncolytic HSV armed with IL-12 elicits local immunity and vaccine therapy towards distant tumors. PLoS Pathog 2018; 14:e1007209. [PMID: 30080893 PMCID: PMC6095629 DOI: 10.1371/journal.ppat.1007209] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 08/16/2018] [Accepted: 07/11/2018] [Indexed: 12/12/2022] Open
Abstract
Oncolytic herpes simplex viruses (oHSVs) showed efficacy in clinical trials and practice. Most of them gain cancer-specificity from deletions/mutations in genes that counteract the host response, and grow selectively in cancer cells defective in anti-viral response. Because of the deletions/mutations, they are frequently attenuated or over-attenuated. We developed next-generation oHSVs, which carry no deletion/mutation, gain cancer-specificity from specific retargeting to tumor cell receptors-e.g. HER2 (human epidermal growth factor receptor 2)-hence are fully-virulent in the targeted cancer cells. The type of immunotherapy they elicit was not predictable, since non-attenuated HSVs induce and then dampen the innate response, whereas deleted/attenuated viruses fail to contrast it, and since the retargeted oHSVs replicate efficiently in tumor cells, but spare other cells in the tumor. We report on the first efficacy study of HER2-retargeted, fully-virulent oHSVs in immunocompetent mice. Their safety profile was very high. Both the unarmed R-LM113 and the IL-12-armed R-115 inhibited the growth of the primary HER2-Lewis lung carcinoma-1 (HER2-LLC1) tumor, R-115 being constantly more efficacious. All the mice that did not die because of the primary treated tumors, were protected from the growth of contralateral untreated tumors. The long-term survivors were protected from a second contralateral tumor, providing additional evidence for an abscopal immunotherapeutic effect. Analysis of the local response highlighted that particularly R-115 unleashed the immunosuppressive tumor microenvironment, i.e. induced immunomodulatory cytokines, including IFNγ, T-bet which promoted Th1 polarization. Some of the tumor infiltrating cells, e.g. CD4+, CD335+ cells were increased in the tumors of all responders mice, irrespective of which virus was employed, whereas CD8+, Foxp3+, CD141+ were increased and CD11b+ cells were decreased preferentially in R-115-treated mice. The durable response included a breakage of tolerance towards both HER2 and the wt tumor cells, and underscored a systemic immunotherapeutic vaccine response.
Collapse
Affiliation(s)
- Valerio Leoni
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Andrea Vannini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Valentina Gatta
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Julie Rambaldi
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Mara Sanapo
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Catia Barboni
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Anna Zaghini
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Patrizia Nanni
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Pier-Luigi Lollini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Costanza Casiraghi
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Gabriella Campadelli-Fiume
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
- * E-mail:
| |
Collapse
|
44
|
Chen AT, Samson BLR, Crupi MJF, Bell JC. Oncolytic viruses: cytolytic agents, replicating immunotherapeutics or both? Future Virol 2018. [DOI: 10.2217/fvl-2018-0047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Ashley T Chen
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, K1H 8L6, Canada
- Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Briana LR Samson
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, K1H 8L6, Canada
- Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Mathieu JF Crupi
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, K1H 8L6, Canada
- Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa K1H 8M5, Canada
| | - John C Bell
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, K1H 8L6, Canada
- Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa K1H 8M5, Canada
| |
Collapse
|
45
|
Martin NT, Bell JC. Oncolytic Virus Combination Therapy: Killing One Bird with Two Stones. Mol Ther 2018; 26:1414-1422. [PMID: 29703699 PMCID: PMC5986726 DOI: 10.1016/j.ymthe.2018.04.001] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 03/29/2018] [Accepted: 04/01/2018] [Indexed: 02/08/2023] Open
Abstract
Over the last 60 years an eclectic collection of microbes has been tested in a variety of pre-clinical models as anti-cancer agents. At the forefront of this research are a number of virus-based platforms that have shown exciting activity in a variety of pre-clinical models and are collectively referred to as oncolytic viruses. Our true understanding of the potential and limitations of this therapeutic modality has been substantially advanced through clinical studies carried out over the last 25 years. Perhaps not surprising, as with all other cancer therapeutics, it has become clear that current oncolytic virus therapeutics on their own are unlikely to be effective in the majority of patients. The greatest therapeutic gains will therefore be made through thoughtful combination strategies built upon an understanding of cancer biology.
Collapse
Affiliation(s)
- Nikolas Tim Martin
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada
| | - John Cameron Bell
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada.
| |
Collapse
|