1
|
Pettas T, Lachanoudi S, Karageorgos FF, Ziogas IA, Fylaktou A, Papalois V, Katsanos G, Antoniadis N, Tsoulfas G. Immunotherapy and liver transplantation for hepatocellular carcinoma: Current and future challenges. World J Transplant 2025; 15:98509. [DOI: 10.5500/wjt.v15.i2.98509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/03/2024] [Accepted: 11/07/2024] [Indexed: 02/21/2025] Open
Abstract
Despite existing curative options like surgical removal, tissue destruction techniques, and liver transplantation for early-stage hepatocellular carcinoma (HCC), the rising incidence and mortality rates of this global health burden necessitate continuous exploration of novel therapeutic strategies. This review critically assesses the dynamic treatment panorama for HCC, focusing specifically on the burgeoning role of immunotherapy in two key contexts: early-stage HCC and downstaging advanced HCC to facilitate liver transplant candidacy. It delves into the unique immunobiology of the liver and HCC, highlighting tumor-mediated immune evasion mechanisms. Analyzing the diverse immunotherapeutic approaches including checkpoint inhibitors, cytokine modulators, vaccines, oncolytic viruses, antigen-targeting antibodies, and adoptive cell therapy, this review acknowledges the limitations of current diagnostic markers alpha-fetoprotein and glypican-3 and emphasizes the need for novel biomarkers for patient selection and treatment monitoring. Exploring the rationale for neoadjuvant and adjuvant immunotherapy in early-stage HCC, current research is actively exploring the safety and effectiveness of diverse immunotherapeutic approaches through ongoing clinical trials. The review further explores the potential benefits and challenges of combining immunotherapy and liver transplant, highlighting the need for careful patient selection, meticulous monitoring, and novel strategies to mitigate post-transplant complications. Finally, this review delves into the latest findings from the clinical research landscape and future directions in HCC management, paving the way for optimizing treatment strategies and improving long-term survival rates for patients with this challenging malignancy.
Collapse
Affiliation(s)
- Theodoros Pettas
- Department of Transplantation Surgery, Center for Research and Innovation in Solid Organ Transplantation, Aristotle University School of Medicine, Thessaloniki 54642, Greece
| | - Sofia Lachanoudi
- Department of Transplantation Surgery, Center for Research and Innovation in Solid Organ Transplantation, Aristotle University School of Medicine, Thessaloniki 54642, Greece
| | - Filippos F Karageorgos
- Department of Transplantation Surgery, Center for Research and Innovation in Solid Organ Transplantation, Aristotle University School of Medicine, Thessaloniki 54642, Greece
| | - Ioannis A Ziogas
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Asimina Fylaktou
- Department of Immunology, National Peripheral Histocompatibility Center, Hippokration General Hospital, Thessaloniki 54642, Greece
| | - Vassilios Papalois
- Department of Transplant Surgery, Imperial College Renal and Transplant Centre, London W12 0HS, United Kingdom
| | - Georgios Katsanos
- Department of Transplantation Surgery, Center for Research and Innovation in Solid Organ Transplantation, Aristotle University of Thessaloniki, School of Medicine, Thessaloniki 54642, Greece
| | - Nikolaos Antoniadis
- Department of Transplantation Surgery, Center for Research and Innovation in Solid Organ Transplantation, Aristotle University School of Medicine, Thessaloniki 54642, Greece
| | - Georgios Tsoulfas
- Department of Transplantation Surgery, Center for Research and Innovation in Solid Organ Transplantation, Aristotle University of Thessaloniki, School of Medicine, Thessaloniki 54642, Greece
| |
Collapse
|
2
|
Chen L, Zuo M, Zhou Q, Wang Y. Oncolytic virotherapy in cancer treatment: challenges and optimization prospects. Front Immunol 2023; 14:1308890. [PMID: 38169820 PMCID: PMC10758479 DOI: 10.3389/fimmu.2023.1308890] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024] Open
Abstract
Oncolytic viruses (OVs) are emerging cancer therapeutics that offer a multifaceted therapeutic platform for the benefits of replicating and lysing tumor cells, being engineered to express transgenes, modulating the tumor microenvironment (TME), and having a tolerable safety profile that does not overlap with other cancer therapeutics. The mechanism of OVs combined with other antitumor agents is based on immune-mediated attack resistance and might benefit patients who fail to achieve durable responses after immune checkpoint inhibitor (ICI) treatment. In this Review, we summarize data on the OV mechanism and limitations of monotherapy, which are currently in the process of combination partner development, especially with ICIs. We discuss some of the hurdles that have limited the preclinical and clinical development of OVs. We also describe the available data and provide guidance for optimizing OVs in clinical practice, as well as a summary of approved and promising novel OVs with clinical indications.
Collapse
Affiliation(s)
- Lingjuan Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Mengsi Zuo
- Department of Oncology, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, Wuhan, China
| | - Qin Zhou
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), College of Bioengineering, Hubei University of Technology, Wuhan, China
| | - Yang Wang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), College of Bioengineering, Hubei University of Technology, Wuhan, China
| |
Collapse
|
3
|
Akram S, Al-Shammari AM, Sahib HB, Jabir MS. Papaverine Enhances the Oncolytic Effects of Newcastle Disease Virus on Breast Cancer In Vitro and In Vivo. Int J Microbiol 2023; 2023:3324247. [PMID: 37720338 PMCID: PMC10504052 DOI: 10.1155/2023/3324247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 10/28/2022] [Accepted: 08/20/2023] [Indexed: 09/19/2023] Open
Abstract
Breast cancer is a lethal disease in females worldwide and needs effective treatment. Targeting cancer cells with selective and safe treatment seems like the best choice, as most chemotherapeutic drugs act unselectively. Papaverine showed promising antitumor activity with a high safety profile and increased blood flow through vasodilation. At the same time, it was widely noticed that virotherapy using the Newcastle disease virus proved to be safe and selective against a broad range of cancer cells. Furthermore, combination therapy is favorable, as it attacks cancer cells with multiple mechanisms and enhances virus entrance into the tumor mass, overcoming cancer cells' resistance to therapy. Therefore, we aimed at assessing the novel combination of the AMHA1 strain of Newcastle disease virus (NDV) and nonnarcotic opium alkaloid (papaverine) against breast cancer models in vitro and in vivo. Methods. In vitro experiments used two human breast cancer cell lines and one normal cell line and were treated with NDV, papaverine, and a combination. The study included a cell viability MTT assay, morphological analysis, and apoptosis detection. Animal experiments used the AN3 mouse mammary adenocarcinoma tumor model. Evaluation of the antitumor activity included growth inhibition measurement; the immunohistochemistry assay measured caspase protein expression. Finally, a semiquantitative microarray assay was used to screen changes in apoptotic proteins. In vitro, results showed that the combination therapy induces synergistic cytotoxicity and apoptosis against cancer cells with a negligible cytotoxic effect on normal cells. In vivo, combination treatment induced a significant antitumor effect with an obvious regression in tumor size and a remarkable and significant expression of caspase-3, caspase-8, and caspase-9 compared to monotherapies. Microarray analysis shows higher apoptosis protein levels in the combination therapy group. In conclusion, this study demonstrated the role of papaverine in enhancing the antitumor activity of NDV, suggesting a promising strategy for breast cancer therapy through nonchemotherapeutic drugs.
Collapse
Affiliation(s)
- Sura Akram
- Department of Pharmacology, College of Medicine, Al-Nahrain University, Baghdad, Iraq
| | - Ahmed Majeed Al-Shammari
- Experimental Therapy, Iraqi Center for Cancer and Medical Genetics Research, Mustansiriyah University, Baghdad, Iraq
| | - Hayder B. Sahib
- Department of Pharmacology, College of Pharmacy, Al-Nahrain University, Baghdad, Iraq
| | - Majid Sakhi Jabir
- Department of Applied Science, University of Technology, Baghdad, Iraq
| |
Collapse
|
4
|
Mori Y, Nishikawa SG, Fratiloiu AR, Tsutsui M, Kataoka H, Joh T, Johnston RN. Modulation of Reoviral Cytolysis (I): Combination Therapeutics. Viruses 2023; 15:1472. [PMID: 37515160 PMCID: PMC10385176 DOI: 10.3390/v15071472] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/17/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
Patients with stage IV gastric cancer suffer from dismal outcomes, a challenge especially in many Asian populations and for which new therapeutic options are needed. To explore this issue, we used oncolytic reovirus in combination with currently used chemotherapeutic drugs (irinotecan, paclitaxel, and docetaxel) for the treatment of gastric and other gastrointestinal cancer cells in vitro and in a mouse model. Cell viability in vitro was quantified by WST-1 assays in human cancer cell lines treated with reovirus and/or chemotherapeutic agents. The expression of reovirus protein and caspase activity was determined by flow cytometry. For in vivo studies, athymic mice received intratumoral injections of reovirus in combination with irinotecan or paclitaxel, after which tumor size was monitored. In contrast to expectations, we found that reoviral oncolysis was only poorly correlated with Ras pathway activation. Even so, the combination of reovirus with chemotherapeutic agents showed synergistic cytopathic effects in vitro, plus enhanced reovirus replication and apoptosis. In vivo experiments showed that reovirus alone can reduce tumor size and that the combination of reovirus with chemotherapeutic agents enhances this effect. Thus, we find that oncolytic reovirus therapy is effective against gastric cancer. Moreover, the combination of reovirus and chemotherapeutic agents synergistically enhanced cytotoxicity in human gastric cancer cell lines in vitro and in vivo. Our data support the use of reovirus in combination with chemotherapy in further clinical trials, and highlight the need for better biomarkers for reoviral oncolytic responsiveness.
Collapse
Affiliation(s)
- Yoshinori Mori
- Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
- Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan
| | - Sandra G Nishikawa
- Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Andreea R Fratiloiu
- Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Mio Tsutsui
- Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Hiromi Kataoka
- Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan
| | - Takashi Joh
- Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan
| | - Randal N Johnston
- Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
5
|
Fukuhara H, Sato YT, Hou J, Iwai M, Todo T. Fusion peptide is superior to co-expressing subunits for arming oncolytic herpes virus with interleukin 12. COMMUNICATIONS MEDICINE 2023; 3:40. [PMID: 36966232 PMCID: PMC10039936 DOI: 10.1038/s43856-023-00270-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 03/06/2023] [Indexed: 03/27/2023] Open
Abstract
BACKGROUND G47∆ is a triple-mutated oncolytic herpes simplex virus type 1 (HSV-1) recently approved as a new drug for malignant glioma in Japan. As the next-generation, we develop armed oncolytic HSV-1 using G47∆ as the backbone. Because oncolytic HSV-1 elicits specific antitumor immunity, interleukin 12 (IL-12) can function as an effective payload to enhance the efficacy. METHODS We evaluate the optimal methods for expressing IL-12 as a payload for G47∆-based oncolytic HSV-1. Two new armed viruses are generated for evaluation by employing different methods to express IL-12: T-mfIL12 expresses murine IL-12 as a fusion peptide, with the genes of two subunits (p35 and p40) linked by bovine elastin motifs, and T-mIL12-IRES co-expresses the subunits, with the two genes separated by an internal ribosome entry site (IRES) sequence. RESULTS T-mfIL12 is significantly more efficient in producing IL-12 than T-mIL12-IRES in all cell lines tested, whereas the expression methods do not affect the replication capabilities and cytopathic effects. In two syngeneic mouse subcutaneous tumor models of Neuro2a and TRAMP-C2, T-mfIL12 exhibits a significantly higher efficacy than T-mIL12-IRES when inoculated intratumorally. Furthermore, T-mfIL12 shows a significantly higher intratumoral expression of functional IL-12, causing stronger stimulation of specific antitumor immune responses than T-mIL12-IRES. CONCLUSIONS The results implicate that a fusion-type expression of IL-12 is a method superior to co-expression of separate subunits, due to higher production of functional IL-12 molecules. This study led to the creation of triple-mutated oncolytic HSV-1 armed with human IL-12 currently used in phase 1/2 trial for malignant melanoma.
Collapse
Affiliation(s)
- Hiroshi Fukuhara
- Division of Innovative Cancer Therapy, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
- Department of Urology, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo, 181-8611, Japan
| | - Yuzuri Tsurumaki Sato
- Division of Innovative Cancer Therapy, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Jiangang Hou
- Division of Innovative Cancer Therapy, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Miwako Iwai
- Division of Innovative Cancer Therapy, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Tomoki Todo
- Division of Innovative Cancer Therapy, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan.
| |
Collapse
|
6
|
Luo YZ, Zhu H. Immunotherapy for advanced or recurrent hepatocellular carcinoma. World J Gastrointest Oncol 2023; 15:405-424. [PMID: 37009314 PMCID: PMC10052663 DOI: 10.4251/wjgo.v15.i3.405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 02/11/2023] [Accepted: 02/28/2023] [Indexed: 03/14/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is associated with high morbidity and mortality, and is prone to intra- and extrahepatic metastasis due to the anatomical and functional characteristics of the liver. Due to the complexity and high relapse rate associated with radical surgery or radiofrequency ablation, immune checkpoint inhibitors (ICIs) are increasingly being used to treat HCC. Several immunotherapeutic agents, along with their combinations, have been clinically approved to treat advanced or recurrent HCC. This review discusses the leading ICIs in practice and those currently undergoing randomized phase 1–3 trials as monotherapy or combination therapy. Furthermore, we summarize the rapidly developing alternative strategies such as chimeric antigen receptor-engineered T cell therapy and tumor vaccines. Combination therapy is a promising potential treatment option. These immunotherapies are also summarized in this review, which provides insights into the advantages, limitations, and novel angles for future research in establishing viable and alternative therapies against HCC.
Collapse
Affiliation(s)
- Ying-Zhe Luo
- Department of Medical Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| | - Hong Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
7
|
Huang CH, Dong T, Phung AT, Shah JR, Larson C, Sanchez AB, Blair SL, Oronsky B, Trogler WC, Reid T, Kummel AC. Full Remission of CAR-Deficient Tumors by DOTAP-Folate Liposome Encapsulation of Adenovirus. ACS Biomater Sci Eng 2022; 8:5199-5209. [PMID: 36395425 DOI: 10.1021/acsbiomaterials.2c00966] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Adenovirus (Ad)-based vectors have shown considerable promise for gene therapy. However, Ad requires the coxsackievirus and adenovirus receptor (CAR) to enter cells efficiently and low CAR expression is found in many human cancers, which hinder adenoviral gene therapies. Here, cationic 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP)-folate liposomes (Df) encapsulating replication-deficient Ad were synthesized, which showed improved transfection efficiency in various CAR-deficient cell lines, including epithelial and hematopoietic cell types. When encapsulating replication-competent oncolytic Ad (TAV255) in DOTAP-folate liposome (TAV255-Df), the adenoviral structural protein, hexon, was readily produced in CAR-deficient cells, and the tumor cell killing ability was 5× higher than that of the non-encapsulated Ad. In CAR-deficient CT26 colon carcinoma murine models, replication-competent TAV255-Df treatment of subcutaneous tumors by intratumoral injection resulted in 67% full tumor remission, prolonged survival, and anti-cancer immunity when mice were rechallenged with cancer cells with no further treatment. The preclinical data shows that DOTAP-folate liposomes could significantly enhance the transfection efficiency of Ad in CAR-deficient cells and, therefore, could be a feasible strategy for applications in cancer treatment.
Collapse
Affiliation(s)
- Ching-Hsin Huang
- Moores Cancer Center, University of California San Diego, 3855 Health Sciences Drive, La Jolla, California 92037, United States
| | - Tao Dong
- Moores Cancer Center, University of California San Diego, 3855 Health Sciences Drive, La Jolla, California 92037, United States
| | - Abraham T Phung
- Moores Cancer Center, University of California San Diego, 3855 Health Sciences Drive, La Jolla, California 92037, United States
| | - Jaimin R Shah
- Moores Cancer Center, University of California San Diego, 3855 Health Sciences Drive, La Jolla, California 92037, United States
| | - Christopher Larson
- EpicentRx, Inc., 11099 North Torrey Pines Road, Suite 160, La Jolla, California 92037, United States
| | - Ana B Sanchez
- EpicentRx, Inc., 11099 North Torrey Pines Road, Suite 160, La Jolla, California 92037, United States
| | - Sarah L Blair
- Moores Cancer Center, University of California San Diego, 3855 Health Sciences Drive, La Jolla, California 92037, United States
| | - Bryan Oronsky
- EpicentRx, Inc., 11099 North Torrey Pines Road, Suite 160, La Jolla, California 92037, United States
| | - William C Trogler
- Department of Chemistry & Biochemistry, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Tony Reid
- EpicentRx, Inc., 11099 North Torrey Pines Road, Suite 160, La Jolla, California 92037, United States
| | - Andrew C Kummel
- Department of Chemistry & Biochemistry, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| |
Collapse
|
8
|
Cassese G, Han HS, Lee B, Lee HW, Cho JY, Panaro F, Troisi RI. Immunotherapy for hepatocellular carcinoma: A promising therapeutic option for advanced disease. World J Hepatol 2022; 14:1862-1874. [PMID: 36340753 PMCID: PMC9627435 DOI: 10.4254/wjh.v14.i10.1862] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/20/2022] [Accepted: 10/03/2022] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related deaths worldwide, and its incidence continues to increase. Despite improvements in both medical and surgical therapies, HCC remains associated with poor outcomes due to its high rates of recurrence and mortality. Approximately 50% of patients require systemic therapies that traditionally consist of tyrosine kinase inhibitors. Recently, however, immune checkpoint inhibitors have revolutionized HCC management, providing new therapeutic options. Despite these major advances, the different factors involved in poor clinical responses and molecular pathways leading to resistance following use of these therapies remain unclear. Alternative strategies, such as adoptive T cell transfer, vaccination, and virotherapy, are currently under evaluation. Combinations of immunotherapies with other systemic or local treatments are also being investigated and may be the most promising opportunities for HCC treatment. The aim of this review is to provide updated information on currently available immunotherapies for HCC as well as future perspectives.
Collapse
Affiliation(s)
- Gianluca Cassese
- Department of Clinical Medicine and Surgery, Division of Minimally Invasive and Robotic HPB Surgery, Federico II University, Naples 80131, Italy
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam 13620, South Korea
| | - Ho-Seong Han
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam 13620, South Korea.
| | - Boram Lee
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam 13620, South Korea
| | - Hae Won Lee
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam 13620, South Korea
| | - Jai Young Cho
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam 13620, South Korea
| | - Fabrizio Panaro
- Department of Surgery, Division of HBP Surgery and Transplantation, Montpellier University Hospital - School of Medicine, Montpellier 34000, France
| | - Roberto Ivan Troisi
- Department of Clinical Medicine and Surgery, Division of Minimally Invasive and Robotic HPB Surgery, Federico II University, Naples 80131, Italy
| |
Collapse
|
9
|
Obaid QA, Al-Shammari AM, Khudair KK. Glucose Deprivation Induced by Acarbose and Oncolytic Newcastle Disease Virus Promote Metabolic Oxidative Stress and Cell Death in a Breast Cancer Model. Front Mol Biosci 2022; 9:816510. [PMID: 35936786 PMCID: PMC9354800 DOI: 10.3389/fmolb.2022.816510] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Cancer cells are distinguished by enhanced glucose uptake and an aerobic glycolysis pathway in which its products support metabolic demands for cancer cell growth and proliferation. Inhibition of aerobic glycolysis is a smart therapeutic approach to target the progression of the cancer cell. We employed acarbose (ACA), a particular alpha-glucosidase inhibitor, to induce glucose deprivation combined with oncolytic Newcastle disease virus (NDV) to enhance antitumor activity. In this work, we used a mouse model of breast cancer with mammary adenocarcinoma tumor cells (AN3) that were treated with ACA, NDV, and a combination of both. The study included antitumor efficacy, relative body weight, glucose level, hexokinase (HK-1) level by ELISA, glycolysis product (pyruvate), total ATP, oxidative stress (ROS and reduced glutathione), and apoptosis by immunohistochemistry. The results showed significant antitumor efficacy against breast cancer after treatment with combination therapy. Antitumor efficacy was accompanied by a reduction in body weight and glucose level, HK-1 downregulation, inhibition of glycolysis products (pyruvate), total ATP, induction of oxidative stress (increase ROS and decrease reduced glutathione), and apoptotic cell death. The findings propose a novel anti–breast cancer combination involving the suppression of glycolysis, glucose deprivation, oxidative stress, and apoptosis, which can be translated clinically.
Collapse
Affiliation(s)
- Qayssar A. Obaid
- Department of Animal Production, College of Agriculture, University of Sumer, Dhi Qar, Iraq
| | - Ahmed Majeed Al-Shammari
- Department of Experimental Therapy, Iraqi Centre for Cancer and Medical Genetic Research, Mustansiriyah University, Baghdad, Iraq
- *Correspondence: Ahmed Majeed Al-Shammari,
| | - Khalisa K. Khudair
- Department of Physiology and Pharmacology, College of Veterinary Medicine/Baghdad University, Baghdad, Iraq
| |
Collapse
|
10
|
Vazeh H, Behboudi E, Hashemzadeh-Omran A, Moradi A. Live-attenuated poliovirus-induced extrinsic apoptosis through Caspase 8 within breast cancer cell lines expressing CD155. Breast Cancer 2022; 29:899-907. [PMID: 35641853 DOI: 10.1007/s12282-022-01372-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 05/06/2022] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Breast cancer is one of the most common cancers among women in the world. Different therapeutic strategies such as radiotherapy, chemotherapy and surgery have been used either individually or in combination. Oncolytic virotherapy is a rising treatment methodology, which utilizes replicating viruses to eliminate tumor cells. The aim of this study was to investigate the oncolytic activity of live-attenuated poliovirus in breast cancer cell lines. MATERIALS AND METHODS The CD155 expression level in two human breast cancer cell lines and a normal breast cell line were evaluated using real-time PCR and flow cytometry. Virus titration was assessed by TCID50. The cytotoxicity of poliovirus on cell line and apoptosis response was investigated by MTT and Caspase 8 and Caspase 9 ELISA kits, respectively. RESULTS This study showed that CD155 gene was expressed significantly (p = 0.001) higher in both human breast cancer cell lines compared to the normal cell line. The protein expression level of CD155 was 98.1%, 96.7%, in MDA_MB231 and MCF_7 cell lines, respectively, whereas the CD155 expression level was 1.3% in MCF_10A. The cytopathic effect of poliovirus in breast cancer cell lines was significantly higher than normal cells (p < 0.05). Extrinsic apoptosis response was more effective than intrinsic apoptosis in both breast cancer cell lines (p < 0.05). CONCLUSION In summary, administration of live-attenuated poliovirus can be a promising treatment to breast cancer. However, in vitro and in vivo studies will be required to evaluate the safety of this strategy.
Collapse
Affiliation(s)
- Hossein Vazeh
- Department of Microbiology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Emad Behboudi
- Department of Microbiology, Golestan University of Medical Sciences, Gorgan, Iran
| | | | - Abdolvahab Moradi
- Department of Microbiology, Golestan University of Medical Sciences, Gorgan, Iran.
| |
Collapse
|
11
|
Carter ME, Hartkopf AD, Wagner A, Volmer LL, Brucker SY, Berchtold S, Lauer UM, Koch A. A Three-Dimensional Organoid Model of Primary Breast Cancer to Investigate the Effects of Oncolytic Virotherapy. Front Mol Biosci 2022; 9:826302. [PMID: 35223990 PMCID: PMC8874275 DOI: 10.3389/fmolb.2022.826302] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/19/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Although several oncolytic viruses have already been tested in early-stage clinical studies of breast cancer, there is still an urgent need to develop patient-derived experimental systems that mimic the response of breast cancer to oncolytic agents in preparation of testing different oncolytic viruses in clinical trials. We addressed this need by developing a protocol to study the effects of oncolytic viruses in stable organoid cell cultures derived from breast cancer tissue.Methods: We used an established three-dimensional organoid model derived from tissue of 10 patients with primary breast cancer. We developed an experimental protocol for infecting organoid cultures with oncolytic viruses and compared the oncolytic effects of a measles vaccine virus (MeV) and a vaccinia virus (GLV) genetically engineered to express either green fluorescent protein (MeV-GFP) and red fluorescent protein (GLV-0b347), respectively, or a suicide gene encoding a fusion of cytosine deaminase with uracil phosphoribosyltransferase (MeV-SCD and GLV-1h94, respectively), thereby enabling enzymatic conversion of the prodrug 5-fluorocytosine (5-FC) into cytotoxic compounds 5-fluorouracil (5-FU) and 5-fluorouridine monophosphate (5-FUMP).Results: The method demonstrated that all oncolytic viruses significantly inhibited cell viability in organoid cultures derived from breast cancer tissue. The oncolytic effects of the oncolytic viruses expressing suicide genes (MeV-SCD and GLV-1h94) were further enhanced by virus-triggered conversion of the prodrug 5-FC to toxic 5-FU and toxic 5-FUMP.Conclusions: We were able to develop a protocol to assess the effects of two different types of oncolytic viruses in stable organoid cell cultures derived from breast cancer tissue. The greatest oncolytic effects were observed when the oncolytic viruses were engineered to express a suicide gene (MeV-SCD and GLV-1h94) in the presence of the prodrug 5-FC. The model therefore provides a promising in vitro method to help further testing and engineering of new generations of virotherapeutic vectors for in vivo use.
Collapse
Affiliation(s)
- Mary E. Carter
- Department of Obstetrics and Gynaecology, University of Tuebingen, Tuebingen, Germany
| | - Andreas D. Hartkopf
- Department of Obstetrics and Gynaecology, University of Tuebingen, Tuebingen, Germany
| | - Anna Wagner
- Department of Obstetrics and Gynaecology, University of Tuebingen, Tuebingen, Germany
| | - Léa L. Volmer
- Department of Obstetrics and Gynaecology, University of Tuebingen, Tuebingen, Germany
| | - Sara Y. Brucker
- Department of Obstetrics and Gynaecology, University of Tuebingen, Tuebingen, Germany
| | - Susanne Berchtold
- Department of Internal Medicine VIII, Medical Oncology and Pneumology, University of Tuebingen, Tuebingen, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Tuebingen, Tuebingen, Germany
| | - Ulrich M. Lauer
- Department of Internal Medicine VIII, Medical Oncology and Pneumology, University of Tuebingen, Tuebingen, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Tuebingen, Tuebingen, Germany
| | - André Koch
- Department of Obstetrics and Gynaecology, University of Tuebingen, Tuebingen, Germany
- *Correspondence: André Koch,
| |
Collapse
|
12
|
Andretta E, Costa C, Longobardi C, Damiano S, Giordano A, Pagnini F, Montagnaro S, Quintiliani M, Lauritano C, Ciarcia R. Potential Approaches Versus Approved or Developing Chronic Myeloid Leukemia Therapy. Front Oncol 2022; 11:801779. [PMID: 34993151 PMCID: PMC8724906 DOI: 10.3389/fonc.2021.801779] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/12/2021] [Indexed: 12/22/2022] Open
Abstract
Tyrosine kinase inhibitors (TKIs) have revolutionized the treatment of patients with chronic myeloid leukemia (CML). However, continued use of these inhibitors has contributed to the increase in clinical resistance and the persistence of resistant leukemic stem cells (LSCs). So, there is an urgent need to introduce additional targeted and selective therapies to eradicate quiescent LSCs, and to avoid the relapse and disease progression. Here, we focused on emerging BCR-ABL targeted and non-BCR-ABL targeted drugs employed in clinical trials and on alternative CML treatments, including antioxidants, oncolytic virus, engineered exosomes, and natural products obtained from marine organisms that could pave the way for new therapeutic approaches for CML patients.
Collapse
Affiliation(s)
- Emanuela Andretta
- Department of Veterinary Medicine and Animal Productions, University of Naples "Federico II", Naples, Italy
| | - Caterina Costa
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Consiglia Longobardi
- Department of Mental, Physical Health and Preventive Medicine, University of Campania "Luigi Vanvitelli", Largo Madonna delle Grazie, Naples, Italy
| | - Sara Damiano
- Department of Veterinary Medicine and Animal Productions, University of Naples "Federico II", Naples, Italy
| | - Antonio Giordano
- Department of Medical Biotechnologies, University of Siena, Siena, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, United States
| | - Francesco Pagnini
- Unit of Radiology, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Serena Montagnaro
- Department of Veterinary Medicine and Animal Productions, University of Naples "Federico II", Naples, Italy
| | | | - Chiara Lauritano
- Marine Biotechnology Department, Stazione Zoologica Anton Dohrn, Naples, Italy
| | - Roberto Ciarcia
- Department of Veterinary Medicine and Animal Productions, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
13
|
Abd El-Hamid BN, Khalil IA, Harashima H. Viral Gene Delivery. THE ADME ENCYCLOPEDIA 2022:1183-1192. [DOI: 10.1007/978-3-030-84860-6_117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
14
|
The Oncolytic Caprine Herpesvirus 1 (CpHV-1) Induces Apoptosis and Synergizes with Cisplatin in Mesothelioma Cell Lines: A New Potential Virotherapy Approach. Viruses 2021; 13:v13122458. [PMID: 34960727 PMCID: PMC8703924 DOI: 10.3390/v13122458] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/12/2021] [Accepted: 12/03/2021] [Indexed: 12/17/2022] Open
Abstract
Malignant mesothelioma (MM) is an aggressive asbestos-related cancer, against which no curative modalities exist. Oncolytic virotherapy is a promising therapeutic approach, for which MM is an ideal candidate; indeed, the pleural location provides direct access for the intra-tumoral injection of oncolytic viruses (OVs). Some non-human OVs offer advantages over human OVs, including the non-pathogenicity in humans and the absence of pre-existing immunity. We previously showed that caprine herpesvirus 1 (CpHV-1), a non-pathogenic virus for humans, can kill different human cancer cell lines. Here, we assessed CpHV-1 effects on MM (NCI-H28, MSTO, NCI-H2052) and non-tumor mesothelial (MET-5A) cells. We found that CpHV-1 reduced cell viability and clonogenic potential in all MM cell lines without affecting non-tumor cells, in which, indeed, we did not detect intracellular viral DNA after treatment. In particular, CpHV-1 induced MM cell apoptosis and accumulation in G0/G1 or S cell cycle phases. Moreover, CpHV-1 strongly synergized with cisplatin, the drug currently used in MM chemotherapy, and this agent combination did not affect normal mesothelial cells. Although further studies are required to elucidate the mechanisms underlying the selective CpHV-1 action on MM cells, our data suggest that the CpHV-1-cisplatin combination could be a feasible strategy against MM.
Collapse
|
15
|
Spiesschaert B, Angerer K, Park J, Wollmann G. Combining Oncolytic Viruses and Small Molecule Therapeutics: Mutual Benefits. Cancers (Basel) 2021; 13:3386. [PMID: 34298601 PMCID: PMC8306439 DOI: 10.3390/cancers13143386] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 02/07/2023] Open
Abstract
The focus of treating cancer with oncolytic viruses (OVs) has increasingly shifted towards achieving efficacy through the induction and augmentation of an antitumor immune response. However, innate antiviral responses can limit the activity of many OVs within the tumor and several immunosuppressive factors can hamper any subsequent antitumor immune responses. In recent decades, numerous small molecule compounds that either inhibit the immunosuppressive features of tumor cells or antagonize antiviral immunity have been developed and tested for. Here we comprehensively review small molecule compounds that can achieve therapeutic synergy with OVs. We also elaborate on the mechanisms by which these treatments elicit anti-tumor effects as monotherapies and how these complement OV treatment.
Collapse
Affiliation(s)
- Bart Spiesschaert
- Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Medical University Innsbruck, 6020 Innsbruck, Austria; (B.S.); (K.A.)
- Institute of Virology, Medical University Innsbruck, 6020 Innsbruck, Austria
- ViraTherapeutics GmbH, 6063 Rum, Austria
- Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach a.d. Riss, Germany;
| | - Katharina Angerer
- Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Medical University Innsbruck, 6020 Innsbruck, Austria; (B.S.); (K.A.)
- Institute of Virology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - John Park
- Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach a.d. Riss, Germany;
| | - Guido Wollmann
- Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Medical University Innsbruck, 6020 Innsbruck, Austria; (B.S.); (K.A.)
- Institute of Virology, Medical University Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
16
|
Kemler I, Karamched B, Neuhauser C, Dingli D. Quantitative imaging and dynamics of tumor therapy with viruses. FEBS J 2021; 288:6273-6285. [PMID: 34213827 DOI: 10.1111/febs.16102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 06/07/2021] [Accepted: 07/01/2021] [Indexed: 12/27/2022]
Abstract
Cancer therapy remains challenging due to the myriad presentations of the disease and the vast genetic diversity of tumors that continuously evolve and often become resistant to therapy. Viruses can be engineered to specifically infect, replicate, and kill tumor cells (tumor virotherapy). Moreover, the viruses can be "armed" with therapeutic genes to enhance their oncolytic effect. Using viruses to treat cancer is exciting and novel and in principle can be used for a broad variety of tumors. However, the approach is distinctly different from other cancer therapies since success depends on establishment of an infection within the tumor and ongoing propagation of the oncolytic virus within the tumor itself. Therefore, the target itself amplifies the therapy. This introduces complex dynamics especially when the immune system is taken into consideration as well as the physical and other biological barriers to virus growth. Understanding these dynamics not only requires mathematical and computational models but also approaches for the noninvasive monitoring of the virus and tumor populations. In this perspective, we discuss strategies and current results to achieve this important goal of understanding these dynamics in pursuit of optimization of oncolytic virotherapy.
Collapse
Affiliation(s)
- Iris Kemler
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Bhargav Karamched
- Department of Mathematics and Institute of Molecular Biophysics, Florida State University, Tallahassee, FL, USA
| | | | - David Dingli
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA.,Division of Hematology and Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
17
|
Fukuhara H, Takeshima Y, Todo T. Triple-mutated oncolytic herpes virus for treating both fast- and slow-growing tumors. Cancer Sci 2021; 112:3293-3301. [PMID: 34036669 PMCID: PMC8353919 DOI: 10.1111/cas.14981] [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] [Received: 02/08/2021] [Revised: 05/05/2021] [Accepted: 05/11/2021] [Indexed: 11/29/2022] Open
Abstract
Oncolytic virus therapy has emerged as a promising treatment option against cancer. To date, oncolytic viruses have been developed for malignant tumors, but the need for this new therapeutic modality also exists for benign and slow‐growing tumors. G47∆ is an oncolytic herpes simplex virus type 1 (HSV‐1) with an enhanced replication capability highly selective to tumor cells due to genetically engineered, triple mutations in the γ34.5, ICP6 and α47 genes. To create a powerful, but safe oncolytic HSV‐1 that replicates efficiently in tumors regardless of growth speed, we used a bacterial artificial chromosome system that allows a desired promoter to regulate the expression of the ICP6 gene in the G47∆ backbone. Restoration of the ICP6 function in a tumor‐specific manner using the hTERT promoter led to a highly capable oncolytic HSV‐1. T‐hTERT was more efficacious in the slow‐growing OS‐RC‐2 and DU145 tumors than the control viruses, while retaining a high efficacy in the fast‐growing U87MG tumors. The safety features are also retained, as T‐hTERT proved safe when inoculated into the brain of HSV‐1 sensitive A/J mice. This new technology should facilitate the use of oncolytic HSV‐1 for all tumors irrespective of growth speed.
Collapse
Affiliation(s)
- Hiroshi Fukuhara
- Department of Urology, Kyorin University School of Medicine, Tokyo, Japan.,Division of Innovative Cancer Therapy, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yuta Takeshima
- Division of Innovative Cancer Therapy, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Tomoki Todo
- Division of Innovative Cancer Therapy, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
18
|
Ni H, Xue J, Wang F, Sun X, Niu M. Nanomedicine Approach to Immunotherapy of Hepatocellular Carcinoma. J Biomed Nanotechnol 2021; 17:771-792. [PMID: 34082866 DOI: 10.1166/jbn.2021.3055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In recent years, the growing studies focused on the immunotherapy of hepatocellular carcinoma and proved the preclinical and clinical promises of host antitumor immune response. However, there were still various obstacles in meeting satisfactory clinic need, such as low response rate, primary resistance and secondary resistance to immunotherapy. Tackling these barriers required a deeper understanding of immune underpinnings and a broader understanding of advanced technology. This review described immune microenvironment of liver and HCC which naturally decided the complexity of immunotherapy, and summarized recent immunotherapy focusing on different points. The ever-growing clues indicated that the instant killing of tumor cell and the subsequent relive of immunosuppressive microenvironment were both indis- pensables. The nanotechnology applied in immunotherapy and the combination with intervention technology was also discussed.
Collapse
Affiliation(s)
- Hongbo Ni
- Department of Interventional Radiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| | - Jian Xue
- Department of Interventional Radiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| | - Fan Wang
- Department of Interventional Radiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| | - Xiaohan Sun
- Department of Interventional Radiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| | - Meng Niu
- Department of Interventional Radiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| |
Collapse
|
19
|
Mozaffari Nejad AS, Noor T, Munim ZH, Alikhani MY, Ghaemi A. A bibliometric review of oncolytic virus research as a novel approach for cancer therapy. Virol J 2021; 18:98. [PMID: 33980264 PMCID: PMC8113799 DOI: 10.1186/s12985-021-01571-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 05/03/2021] [Indexed: 02/06/2023] Open
Abstract
Background In recent years, oncolytic viruses (OVs) have drawn attention as a novel therapy to various types of cancers, both in clinical and preclinical cancer studies all around the world. Consequently, researchers have been actively working on enhancing cancer therapy since the early twentieth century. This study presents a systematic review of the literature on OVs, discusses underlying research clusters and, presents future directions of OVs research. Methods A total of 1626 published articles related to OVs as cancer therapy were obtained from the Web of Science (WoS) database published between January 2000 and March 2020. Various aspects of OVs research, including the countries/territories, institutions, journals, authors, citations, research areas, and content analysis to find trending and emerging topics, were analysed using the bibliometrix package in the R-software. Results In terms of the number of publications, the USA based researchers were the most productive (n = 611) followed by Chinese (n = 197), and Canadian (n = 153) researchers. The Molecular Therapy journal ranked first both in terms of the number of publications (n = 133) and local citations (n = 1384). The most prominent institution was Mayo Clinic from the USA (n = 117) followed by the University of Ottawa from Canada (n = 72), and the University of Helsinki from Finland (n = 63). The most impactful author was Bell J.C with the highest number of articles (n = 67) and total local citations (n = 885). The most impactful article was published in the Cell journal. In addition, the latest OVs research mainly builds on four research clusters. Conclusion The domain of OVs research has increased at a rapid rate from 2000 to 2020. Based on the synthesis of reviewed studies, adenovirus, herpes simplex virus, reovirus, and Newcastle disease virus have shown potent anti-cancer activity. Developed countries such as the USA, Canada, the UK, and Finland were the most productive, hence, contributed most to this field. Further collaboration will help improve the clinical research translation of this therapy and bring benefits to cancer patients worldwide.
Collapse
Affiliation(s)
| | - Tehjeeb Noor
- Faculty of Medicine, University of Bergen, Horten, Norway
| | - Ziaul Haque Munim
- Faculty of Technology, Natural and Maritime Sciences, University of South-Eastern Norway, Horten, Norway
| | - Mohammad Yousef Alikhani
- Department of Microbiology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Amir Ghaemi
- Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
20
|
Jiang B, Huang D, He W, Guo W, Yin X, Forsyth P, Lun X, Wang Z. Inhibition of glioma using a novel non-neurotoxic vesicular stomatitis virus. Neurosurg Focus 2021; 50:E9. [PMID: 33524950 DOI: 10.3171/2020.11.focus20839] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/17/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The aim of this study was to demonstrate the in vivo safety and antitumor effect of a novel recombinant vesicular stomatitis virus (VSV): G protein less (GLESS)-fusion-associated small transmembrane (FAST)-VSV. METHODS Viral infection efficiency and cell proliferation were detected using an inverted fluorescence microscope and alarmaBlue assay, respectively. To evaluate the safety of the virus, different doses of GLESS-FAST-VSV and a positive control virus (VSV∆M51) were injected into normal F344 rats and C57BL/6 mice, and each animal's weight, survival time, and pathological changes were examined on the following day. To evaluate the efficacy of the virus, RG2 and GL261 cells were used to construct rat and mouse glioma models, respectively, via a stereotactic method. After multiple intratumoral injections of the virus, tumor growth (size) and the survival time of the animals were observed. RESULTS In vitro experiments showed that GLESS-FAST-VSV could infect and kill brain tumor cells and had less toxic effects on normal cells. After direct injection of GLESS-FAST-VSV into the animal brains, all animals tolerated the virus well, and no animal death, encephalitis, or ventriculitis was observed. In contrast, all animals that received brain injections of VSV∆M51 in the brain died. Moreover, multiple injections of GLESS-FAST-VSV in brain tumors significantly prolonged the survival of normal-immunity animals harboring brain tumors. CONCLUSIONS GLESS-FAST-VSV exhibited little neurotoxicity and could be injected directly into the tumor to effectively inhibit tumor growth and prolong the survival of normal-immunity animals, laying a theoretical foundation for the early application of such viruses in clinical trials.
Collapse
Affiliation(s)
- Bin Jiang
- 1Department of Neurosurgery, Qilu Hospital of Shandong University, Qingdao, Shandong, China; and
| | - Dezhang Huang
- 1Department of Neurosurgery, Qilu Hospital of Shandong University, Qingdao, Shandong, China; and
| | - Wei He
- 1Department of Neurosurgery, Qilu Hospital of Shandong University, Qingdao, Shandong, China; and
| | | | -
- 1Department of Neurosurgery, Qilu Hospital of Shandong University, Qingdao, Shandong, China; and
| | | | -
- 1Department of Neurosurgery, Qilu Hospital of Shandong University, Qingdao, Shandong, China; and
| | - Peter Forsyth
- 2Department of Oncology, University of Calgary, Southern Alberta Cancer Research Institute, Calgary, Alberta, Canada
| | - Xueqing Lun
- 2Department of Oncology, University of Calgary, Southern Alberta Cancer Research Institute, Calgary, Alberta, Canada
| | - Zhigang Wang
- 1Department of Neurosurgery, Qilu Hospital of Shandong University, Qingdao, Shandong, China; and
| |
Collapse
|
21
|
Béguin J, Gantzer M, Farine I, Foloppe J, Klonjkowski B, Maurey C, Quéméneur É, Erbs P. Safety, biodistribution and viral shedding of oncolytic vaccinia virus TG6002 administered intravenously in healthy beagle dogs. Sci Rep 2021; 11:2209. [PMID: 33500518 PMCID: PMC7838210 DOI: 10.1038/s41598-021-81831-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/11/2021] [Indexed: 01/30/2023] Open
Abstract
Oncolytic virotherapy is an emerging strategy that uses replication-competent viruses to kill tumor cells. We have reported the oncolytic effects of TG6002, a recombinant oncolytic vaccinia virus, in preclinical human xenograft models and canine tumor explants. To assess the safety, biodistribution and shedding of TG6002 administered by the intravenous route, we conducted a study in immune-competent healthy dogs. Three dogs each received a single intravenous injection of TG6002 at 105 PFU/kg, 106 PFU/kg or 107 PFU/kg, and one dog received three intravenous injections at 107 PFU/kg. The injections were well tolerated without any clinical, hematological or biochemical adverse events. Viral genomes were only detected in blood at the earliest sampling time point of one-hour post-injection at 107 PFU/kg. Post mortem analyses at day 35 allowed detection of viral DNA in the spleen of the dog which received three injections at 107 PFU/kg. Viral genomes were not detected in the urine, saliva or feces of any dogs. Seven days after the injections, a dose-dependent antibody mediated immune response was identified. In conclusion, intravenous administration of TG6002 shows a good safety profile, supporting the initiation of clinical trials in canine cancer patients as well as further development as a human cancer therapy.
Collapse
Affiliation(s)
- Jérémy Béguin
- Transgene, Illkirch-Graffenstaden, France
- UMR Virologie, INRA, Ecole Nationale Vétérinaire d'Alfort, ANSES, Université Paris-Est, Maisons-Alfort, France
- Department of Internal Medicine, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, Maisons-Alfort, France
| | | | | | | | - Bernard Klonjkowski
- UMR Virologie, INRA, Ecole Nationale Vétérinaire d'Alfort, ANSES, Université Paris-Est, Maisons-Alfort, France
| | - Christelle Maurey
- Department of Internal Medicine, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, Maisons-Alfort, France
| | | | | |
Collapse
|
22
|
Abd El-Hamid BN, Khalil IA, Harashima H. Viral Gene Delivery. THE ADME ENCYCLOPEDIA 2021:1-10. [DOI: 10.1007/978-3-030-51519-5_117-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 02/01/2021] [Indexed: 09/01/2023]
|
23
|
Al-Ziaydi AG, Al-Shammari AM, Hamzah MI, kadhim HS, Jabir MS. Hexokinase inhibition using D-Mannoheptulose enhances oncolytic newcastle disease virus-mediated killing of breast cancer cells. Cancer Cell Int 2020; 20:420. [DOI: https:/doi.org/10.1186/s12935-020-01514-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 08/24/2020] [Indexed: 01/17/2025] Open
Abstract
Abstract
Background
Most cancer cells exhibit increased glycolysis and use this metabolic pathway cell growth and proliferation. Targeting cancer cells’ metabolism is a promising strategy in inhibiting cancer cell progression. We used D-Mannoheptulose, a specific hexokinase inhibitor, to inhibit glycolysis to enhance the Newcastle disease virus anti-tumor effect.
Methods
Human breast cancer cells were treated by NDV and/or hexokinase inhibitor. The study included cell viability, apoptosis, and study levels of hexokinase enzyme, pyruvate, ATP, and acidity. The combination index was measured to determine the synergism of NDV and hexokinase inhibitor.
Results
The results showed synergistic cytotoxicity against breast cancer cells by combination therapy but no cytotoxic effect against normal cells. The effect was accompanied by apoptotic cell death and hexokinase downregulation and inhibition to glycolysis products, pyruvate, ATP, and acidity.
Conclusions
The combination treatment showed safe significant tumor cell proliferation inhibition compared to monotherapies suggesting a novel strategy for anti-breast cancer therapy through glycolysis inhibition by hexokinase downregulation.
Collapse
|
24
|
Zhang B, Cheng P. Improving antitumor efficacy via combinatorial regimens of oncolytic virotherapy. Mol Cancer 2020; 19:158. [PMID: 33172438 PMCID: PMC7656670 DOI: 10.1186/s12943-020-01275-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/22/2020] [Indexed: 02/07/2023] Open
Abstract
As a promising therapeutic strategy, oncolytic virotherapy has shown potent anticancer efficacy in numerous pre-clinical and clinical trials. Oncolytic viruses have the capacity for conditional-replication within carcinoma cells leading to cell death via multiple mechanisms, including direct lysis of neoplasms, induction of immunogenic cell death, and elicitation of innate and adaptive immunity. In addition, these viruses can be engineered to express cytokines or chemokines to alter tumor microenvironments. Combination of oncolytic virotherapy with other antitumor therapeutic modalities, such as chemotherapy and radiation therapy as well as cancer immunotherapy can be used to target a wider range of tumors and promote therapeutic efficacy. In this review, we outline the basic biological characteristics of oncolytic viruses and the underlying mechanisms that support their use as promising antitumor drugs. We also describe the enhanced efficacy attributed to virotherapy combined with other drugs for the treatment of cancer.
Collapse
Affiliation(s)
- Bin Zhang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu, 610041, PR China
| | - Ping Cheng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu, 610041, PR China.
| |
Collapse
|
25
|
Current Perspectives on Therapies, Including Drug Delivery Systems, for Managing Glioblastoma Multiforme. ACS Chem Neurosci 2020; 11:2962-2977. [PMID: 32945654 DOI: 10.1021/acschemneuro.0c00555] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma multiforme (GBM), a standout among the most dangerous class of central nervous system (CNS) cancer, is most common and is an aggressive malignant brain tumor in adults. In spite of developments in modality therapy, it remains mostly incurable. Consequently, the need for novel systems, strategies, or therapeutic approaches for enhancing the assortment of active agents meant for GBM becomes an important criterion. Currently, cancer research focuses mainly on improving the treatment of GBM via diverse novel drug delivery systems. The treatment options at diagnosis are multimodal and include radiation therapy. Moreover, significant advances in understanding the molecular pathology of GBM and associated cell signaling pathways have opened opportunities for new therapies. Innovative treatment such as immunotherapy also gives hope for enhanced survival. The objective of this work was to collect and report the recent research findings to manage GBM. The present review includes existing novel drug delivery systems and therapies intended for managing GBM. Reported novel drug delivery systems and diverse therapies seem to be precise, secure, and relatively effective, which could lead to a new track for the obliteration of GBM.
Collapse
|
26
|
Kole C, Charalampakis N, Tsakatikas S, Vailas M, Moris D, Gkotsis E, Kykalos S, Karamouzis MV, Schizas D. Immunotherapy for Hepatocellular Carcinoma: A 2021 Update. Cancers (Basel) 2020; 12:2859. [PMID: 33020428 PMCID: PMC7600093 DOI: 10.3390/cancers12102859] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/26/2020] [Accepted: 09/30/2020] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of one of the most frequent liver cancers and the fourth leading cause of cancer-related mortality worldwide. Current treatment options such as surgery, neoadjuvant chemoradiotherapy, liver transplantation, and radiofrequency ablation will benefit only a very small percentage of patients. Immunotherapy is a novel treatment approach representing an effective and promising option against several types of cancer. The aim of our study is to present the currently ongoing clinical trials and to evaluate the efficacy of immunotherapy in HCC. In this paper, we demonstrate that combination of different immunotherapies or immunotherapy with other modalities results in better overall survival (OS) and progression-free survival (PFS) compared to single immunotherapy agent. Another objective of this paper is to demonstrate and highlight the importance of tumor microenvironment as a predictive and prognostic marker and its clinical implications in immunotherapy response.
Collapse
Affiliation(s)
- Christo Kole
- First Department of Surgery, National and Kapodistrian University of Athens, Laikon General Hospital, 115 27 Athens, Greece; (C.K.); (M.V.); (E.G.); (D.S.)
| | - Nikolaos Charalampakis
- Department of Medical Oncology, Metaxa Cancer Hospital, 185 37 Athens, Greece; (N.C.); (S.T.)
| | - Sergios Tsakatikas
- Department of Medical Oncology, Metaxa Cancer Hospital, 185 37 Athens, Greece; (N.C.); (S.T.)
| | - Michail Vailas
- First Department of Surgery, National and Kapodistrian University of Athens, Laikon General Hospital, 115 27 Athens, Greece; (C.K.); (M.V.); (E.G.); (D.S.)
| | - Dimitrios Moris
- Department of Surgery, Duke University School of Medicine, Durham, NC 27707, USA;
| | - Efthymios Gkotsis
- First Department of Surgery, National and Kapodistrian University of Athens, Laikon General Hospital, 115 27 Athens, Greece; (C.K.); (M.V.); (E.G.); (D.S.)
| | - Stylianos Kykalos
- Second Propedeutic Department of Surgery, National and Kapodistrian University of Athens, Laikon General Hospital, 115 27 Athens, Greece;
| | - Michalis V. Karamouzis
- Molecular Oncology Unit, Department of Biological Chemistry, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Dimitrios Schizas
- First Department of Surgery, National and Kapodistrian University of Athens, Laikon General Hospital, 115 27 Athens, Greece; (C.K.); (M.V.); (E.G.); (D.S.)
| |
Collapse
|
27
|
Al-Ziaydi AG, Al-Shammari AM, Hamzah MI, kadhim HS, Jabir MS. Hexokinase inhibition using D-Mannoheptulose enhances oncolytic newcastle disease virus-mediated killing of breast cancer cells. Cancer Cell Int 2020; 20:420. [PMID: 32874134 PMCID: PMC7456035 DOI: 10.1186/s12935-020-01514-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 08/24/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Most cancer cells exhibit increased glycolysis and use this metabolic pathway cell growth and proliferation. Targeting cancer cells' metabolism is a promising strategy in inhibiting cancer cell progression. We used D-Mannoheptulose, a specific hexokinase inhibitor, to inhibit glycolysis to enhance the Newcastle disease virus anti-tumor effect. METHODS Human breast cancer cells were treated by NDV and/or hexokinase inhibitor. The study included cell viability, apoptosis, and study levels of hexokinase enzyme, pyruvate, ATP, and acidity. The combination index was measured to determine the synergism of NDV and hexokinase inhibitor. RESULTS The results showed synergistic cytotoxicity against breast cancer cells by combination therapy but no cytotoxic effect against normal cells. The effect was accompanied by apoptotic cell death and hexokinase downregulation and inhibition to glycolysis products, pyruvate, ATP, and acidity. CONCLUSIONS The combination treatment showed safe significant tumor cell proliferation inhibition compared to monotherapies suggesting a novel strategy for anti-breast cancer therapy through glycolysis inhibition by hexokinase downregulation.
Collapse
Affiliation(s)
- Ahmed Ghdhban Al-Ziaydi
- Department of Medical Chemistry, College of Medicine, University of Al-Qadisiyah, Al Diwaniyah, Iraq
| | - Ahmed Majeed Al-Shammari
- Experimental Therapy, Iraqi Center for Cancer and Medical Genetics Research, Mustansiriyah University, Baghdad, Iraq
| | | | - Haider Sabah kadhim
- Department of Microbiology, College of Medicine, Al-Nahrain University, Baghdad, Iraq
| | - Majid Sakhi Jabir
- Division of Biotechnology, Department of Applied Science, University of Technology, Baghdad, Iraq
| |
Collapse
|
28
|
Deng L, Yang X, Fan J, Ding Y, Peng Y, Xu D, Huang B, Hu Z. IL-24-Armed Oncolytic Vaccinia Virus Exerts Potent Antitumor Effects via Multiple Pathways in Colorectal Cancer. Oncol Res 2020; 28:579-590. [PMID: 32641200 PMCID: PMC7962938 DOI: 10.3727/096504020x15942028641011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Colorectal cancer is an aggressive malignancy for which there are limited treatment options. Oncolytic vaccinia virus is being developed as a novel strategy for cancer therapy. Arming vaccinia virus with immunostimulatory cytokines can enhance the tumor cell-specific replication and antitumor efficacy. Interleukin-24 (IL-24) is an important immune mediator, as well as a broad-spectrum tumor suppressor. We constructed a targeted vaccinia virus of Guang9 strain harboring IL-24 (VG9-IL-24) to evaluate its antitumor effects. In vitro, VG9-IL-24 induced an increased number of apoptotic cells and blocked colorectal cancer cells in the G2/M phase of the cell cycle. VG9-IL-24 induced apoptosis in colorectal cancer cells via multiple apoptotic signaling pathways. In vivo, VG9-IL-24 significantly inhibited the tumor growth and prolonged the survival both in human and murine colorectal cancer models. In addition, VG9-IL-24 stimulated multiple antitumor immune responses and direct bystander antitumor activity. Our results indicate that VG9-IL-24 can inhibit the growth of colorectal cancer tumor by inducing oncolysis and apoptosis as well as stimulating the antitumor immune effects. These findings indicate that VG9-IL-24 may exert a potential therapeutic strategy for combating colorectal cancer.
Collapse
Affiliation(s)
- Lili Deng
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear MedicineWuxiP.R. China
| | - Xue Yang
- Wuxi Childrens Hospital, Wuxi Peoples Hospital Affiliated to Nanjing Medical UniversityWuxiP.R. China
| | - Jun Fan
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear MedicineWuxiP.R. China
| | - Yuedi Ding
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear MedicineWuxiP.R. China
| | - Ying Peng
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear MedicineWuxiP.R. China
| | - Dong Xu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear MedicineWuxiP.R. China
| | - Biao Huang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear MedicineWuxiP.R. China
| | - Zhigang Hu
- Wuxi Childrens Hospital, Wuxi Peoples Hospital Affiliated to Nanjing Medical UniversityWuxiP.R. China
| |
Collapse
|
29
|
Deng L, Yang X, Fan J, Ding Y, Peng Y, Xu D, Huang B, Hu Z. An Oncolytic Vaccinia Virus Armed with GM-CSF and IL-24 Double Genes for Cancer Targeted Therapy. Onco Targets Ther 2020; 13:3535-3544. [PMID: 32425553 PMCID: PMC7196195 DOI: 10.2147/ott.s249816] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/07/2020] [Indexed: 12/28/2022] Open
Abstract
Purpose Targeted oncolytic vaccinia virus is an attractive candidate for cancer therapy due to its replication causing lysis of infected tumor cells as well as a delivery vector to overexpress therapeutic transgenes. This study constructed a novel oncolytic vaccinia virus carrying granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-24 (IL-24) double genes to improve efficacy for cancer therapy. Methods Vaccinia virus co-expressing GM-CSF and IL-24 based on Chinese Guang9 strain (VG9-GMCSF-IL24) was constructed with disruption of the viral thymidine kinase (TK) gene. The cytotoxicity of VG9-GMCSF-IL24 in various cell lines was assessed by MTT. The synergistic antitumor effect of VG9-GMCSF-IL24 in vivo was assessed on multiple tumor models. Results In vitro cytotoxicity assay showed that VG9-GMCSF-IL24 exerted a strongly cytotoxic effect on cancer cells, but with no significant cytotoxicity to normal cells. Significant tumor growth inhibition and prolonged survival were observed in different tumor models treated with VG9-GMCSF-IL24. Additionally, systemic and specific antitumoral immunity was investigated in vivo, and enhanced antitumor immunity was observed in VG9-GMCSF-IL24-treated mice. Conclusion Our results indicated that VG9-mediated GM-CSF and IL-24 co-expression performed cooperative and overlapping antitumor effect. As a novel and effective therapeutic strategy for cancer, the combination of oncolysis and immunotherapy with vaccinia virus carrying one or more immunostimulatory genes may have a satisfactory clinical application prospect.
Collapse
Affiliation(s)
- Lili Deng
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, People's Republic of China
| | - Xue Yang
- Wuxi Children's Hospital, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214023, People's Republic of China
| | - Jun Fan
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, People's Republic of China
| | - Yuedi Ding
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, People's Republic of China
| | - Ying Peng
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, People's Republic of China
| | - Dong Xu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, People's Republic of China
| | - Biao Huang
- School of Life Science, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, People's Republic of China
| | - Zhigang Hu
- Wuxi Children's Hospital, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214023, People's Republic of China
| |
Collapse
|
30
|
Chen SH, Sun JM, Chen BM, Lin SC, Chang HF, Collins S, Chang D, Wu SF, Lu YC, Wang W, Chen TC, Kasahara N, Wang HE, Tai CK. Efficient Prodrug Activator Gene Therapy by Retroviral Replicating Vectors Prolongs Survival in an Immune-Competent Intracerebral Glioma Model. Int J Mol Sci 2020; 21:ijms21041433. [PMID: 32093290 PMCID: PMC7073086 DOI: 10.3390/ijms21041433] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 02/13/2020] [Accepted: 02/19/2020] [Indexed: 12/22/2022] Open
Abstract
Prodrug activator gene therapy mediated by murine leukemia virus (MLV)-based retroviral replicating vectors (RRV) was previously shown to be highly effective in killing glioma cells both in culture and in vivo. To avoid receptor interference and enable dual vector co-infection with MLV-RRV, we have developed another RRV based on gibbon ape leukemia virus (GALV) that also shows robust replicative spread in a wide variety of tumor cells. We evaluated the potential of GALV-based RRV as a cancer therapeutic agent by incorporating yeast cytosine deaminase (CD) and E. coli nitroreductase (NTR) prodrug activator genes into the vector. The expression of CD and NTR genes from GALV-RRV achieved highly efficient delivery of these prodrug activator genes to RG-2 glioma cells, resulting in enhanced cytotoxicity after administering their respective prodrugs 5-fluorocytosine and CB1954 in vitro. In an immune-competent intracerebral RG-2 glioma model, GALV-mediated CD and NTR gene therapy both significantly suppressed tumor growth with CB1954 administration after a single injection of vector supernatant. However, NTR showed greater potency than CD, with control animals receiving GALV-NTR vector alone (i.e., without CB1954 prodrug) showing extensive tumor growth with a median survival time of 17.5 days, while animals receiving GALV-NTR and CB1954 showed significantly prolonged survival with a median survival time of 30 days. In conclusion, GALV-RRV enabled high-efficiency gene transfer and persistent expression of NTR, resulting in efficient cell killing, suppression of tumor growth, and prolonged survival upon CB1954 administration. This validates the use of therapeutic strategies employing this prodrug activator gene to arm GALV-RRV, and opens the door to the possibility of future combination gene therapy with CD-armed MLV-RRV, as the latter vector is currently being evaluated in clinical trials.
Collapse
Affiliation(s)
- Shih-Han Chen
- Section of Neurosurgery, Department of Surgery, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chia-Yi 600, Taiwan; (S.-H.C.); (J.-M.S.)
| | - Jui-Ming Sun
- Section of Neurosurgery, Department of Surgery, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chia-Yi 600, Taiwan; (S.-H.C.); (J.-M.S.)
- Department of Biotechnology, Asia University, Taichung 413, Taiwan
| | - Bing-Mao Chen
- Department of Biomedical Sciences, National Chung Cheng University, Chia-Yi 621, Taiwan; (B.-M.C.); (S.-C.L.); (H.-F.C.); (D.C.); (S.-F.W.)
| | - Sheng-Che Lin
- Department of Biomedical Sciences, National Chung Cheng University, Chia-Yi 621, Taiwan; (B.-M.C.); (S.-C.L.); (H.-F.C.); (D.C.); (S.-F.W.)
| | - Hao-Fang Chang
- Department of Biomedical Sciences, National Chung Cheng University, Chia-Yi 621, Taiwan; (B.-M.C.); (S.-C.L.); (H.-F.C.); (D.C.); (S.-F.W.)
| | - Sara Collins
- Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA; (S.C.); (N.K.)
| | - Deching Chang
- Department of Biomedical Sciences, National Chung Cheng University, Chia-Yi 621, Taiwan; (B.-M.C.); (S.-C.L.); (H.-F.C.); (D.C.); (S.-F.W.)
| | - Shu-Fen Wu
- Department of Biomedical Sciences, National Chung Cheng University, Chia-Yi 621, Taiwan; (B.-M.C.); (S.-C.L.); (H.-F.C.); (D.C.); (S.-F.W.)
| | - Yin-Che Lu
- Department of Health and Nutrition, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan;
| | - Weijun Wang
- Department of Neurosurgery, University of Southern California, Los Angeles, CA 90033, USA; (W.W.); (T.C.C.)
| | - Thomas C. Chen
- Department of Neurosurgery, University of Southern California, Los Angeles, CA 90033, USA; (W.W.); (T.C.C.)
| | - Noriyuki Kasahara
- Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA; (S.C.); (N.K.)
- Department of Radiation Oncology, University of California, San Francisco, CA 94143, USA
| | - Hsin-Ell Wang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei 112, Taiwan
- Correspondence: (H.-E.W.); (C.-K.T.)
| | - Chien-Kuo Tai
- Department of Biomedical Sciences, National Chung Cheng University, Chia-Yi 621, Taiwan; (B.-M.C.); (S.-C.L.); (H.-F.C.); (D.C.); (S.-F.W.)
- Correspondence: (H.-E.W.); (C.-K.T.)
| |
Collapse
|
31
|
Tazawa H, Hasei J, Yano S, Kagawa S, Ozaki T, Fujiwara T. Bone and Soft-Tissue Sarcoma: A New Target for Telomerase-Specific Oncolytic Virotherapy. Cancers (Basel) 2020; 12:cancers12020478. [PMID: 32085583 PMCID: PMC7072448 DOI: 10.3390/cancers12020478] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/10/2020] [Accepted: 02/10/2020] [Indexed: 12/17/2022] Open
Abstract
Adenovirus serotype 5 (Ad5) is widely and frequently used as a virus vector in cancer gene therapy and oncolytic virotherapy. Oncolytic virotherapy is a novel antitumor treatment for inducing lytic cell death in tumor cells without affecting normal cells. Based on the Ad5 genome, we have generated three types of telomerase-specific replication-competent oncolytic adenoviruses: OBP-301 (Telomelysin), green fluorescent protein (GFP)-expressing OBP-401 (TelomeScan), and tumor suppressor p53-armed OBP-702. These viruses drive the expression of the adenoviral E1A and E1B genes under the control of the hTERT (human telomerase reverse transcriptase-encoding gene) promoter, providing tumor-specific virus replication. This review focuses on the therapeutic potential of three hTERT promoter-driven oncolytic adenoviruses against bone and soft-tissue sarcoma cells with telomerase activity. OBP-301 induces the antitumor effect in monotherapy or combination therapy with chemotherapeutic drugs via induction of autophagy and apoptosis. OBP-401 enables visualization of sarcoma cells within normal tissues by serving as a tumor-specific labeling reagent for fluorescence-guided surgery via induction of GFP expression. OBP-702 exhibits a profound antitumor effect in OBP-301-resistant sarcoma cells via activation of the p53 signaling pathway. Taken together, telomerase-specific oncolytic adenoviruses are promising antitumor reagents that are expected to provide novel therapeutic options for the treatment of bone and soft-tissue sarcomas.
Collapse
Affiliation(s)
- Hiroshi Tazawa
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama 700-8558, Japan
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.Y.); (S.K.); (T.F.)
- Correspondence: ; Tel.: +81-86-235-7491; Fax: +81-86-235-7492
| | - Joe Hasei
- Department of Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (J.H.); (T.O.)
| | - Shuya Yano
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.Y.); (S.K.); (T.F.)
| | - Shunsuke Kagawa
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.Y.); (S.K.); (T.F.)
- Minimally Invasive Therapy Center, Okayama University Hospital, Okayama 700-8558, Japan
| | - Toshifumi Ozaki
- Department of Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (J.H.); (T.O.)
| | - Toshiyoshi Fujiwara
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.Y.); (S.K.); (T.F.)
| |
Collapse
|
32
|
Electroactive composite scaffold with locally expressed osteoinductive factor for synergistic bone repair upon electrical stimulation. Biomaterials 2019; 230:119617. [PMID: 31771859 DOI: 10.1016/j.biomaterials.2019.119617] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 10/30/2019] [Accepted: 11/10/2019] [Indexed: 02/05/2023]
Abstract
Tissue engineering is a promising strategy for the repair of large-scale bone defects, in which scaffolds and growth factors are two critical issues influencing the efficacy of bone regeneration. Unfortunately, the broad application of growth factors is limited by their poor stability in the scaffolds. In the present study, the strictly controlled expression of human bone morphogenetic protein-4 (hBMP-4) in the presence of doxycycline is achieved by adding an hBMP-4 gene fragment into a non-viral artificial restructuring plasmid vector (pSTAR) to form the pSTAR-hBMP-4 plasmid (phBMP-4). Furthermore, the controlled release of phBMP-4 is obtained with an electroactive tissue engineering scaffold, generated by combining a triblock copolymer of poly(l-lactic acid)-block-aniline pentamer-block-poly(l-lactic acid) (PLA-AP) with poly(lactic-co-glycolic acid)/hydroxyapatite (PLGA/HA). This PLGA/HA/PLA-AP/phBMP-4 composite scaffold, with controlled gene release and Dox-regulated gene expression upon electrical stimulation, operating synergistically, exhibits an improved cell proliferation ability, enhanced osteogenesis differentiation in vitro, and effective bone healing in vivo in a rabbit radial defect model. Taking these results together, the proposed smart PLGA/HA/PLA-AP/phBMP-4 scaffold lays a solid theoretical and experimental basis for future applications of such multi-functional materials in bone tissue engineering to help patients in need.
Collapse
|
33
|
Al-Shammari AM, Abdullah AH, Allami ZM, Yaseen NY. 2-Deoxyglucose and Newcastle Disease Virus Synergize to Kill Breast Cancer Cells by Inhibition of Glycolysis Pathway Through Glyceraldehyde3-Phosphate Downregulation. Front Mol Biosci 2019; 6:90. [PMID: 31612140 PMCID: PMC6777003 DOI: 10.3389/fmolb.2019.00090] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 09/11/2019] [Indexed: 01/16/2023] Open
Abstract
Targeting cancer cells metabolism is promising strategy in inhibiting cancer cells progression that are known to exhibit increased aerobic glycolysis. We used the glucose analog 2-Deoxyglucose (2-DG) as a competitor molecule of glucose. To further enhance the effectiveness of 2-DG, the Newcastle disease virus (NDV) was used as a combination virotherapy to enhance the anti-tumor effect. Human and mouse-breast cancer cells were treated by NDV and/or 2-DG. The effect was analyzed by study cell viability, apoptosis and level of glyceraldehyde3-phosphate (GAPDH) by ELISA and QPCR assays. Synergistic cytotoxicity was found after a 72-h treatment of human- and mouse-breast cancer cells with 2-DG in combination with NDV at different concentrations. The synergistic cytotoxicity was accompanied by apoptotic cell death and GAPDH downregulation and inhibition to glycolysis product pyruvate. The combination treatment showed significant tumor growth inhibition compared to single treatments in vivo. Our results suggest the effectiveness of a novel strategy for anti-breast cancer therapy through glycolysis inhibition and GAPDH downregulation.
Collapse
Affiliation(s)
- Ahmed Majeed Al-Shammari
- Experimental Therapy Department, Iraqi Centre for Cancer and Medical Genetic Research, Mustansiriyah University, Baghdad, Iraq
| | - Amer Hasan Abdullah
- Experimental Therapy Department, Iraqi Centre for Cancer and Medical Genetic Research, Mustansiriyah University, Baghdad, Iraq
| | - Zainab Majid Allami
- Department of Chemistry, College of Science, Mustansiriyah University, Baghdad, Iraq
| | - Nahi Y Yaseen
- Experimental Therapy Department, Iraqi Centre for Cancer and Medical Genetic Research, Mustansiriyah University, Baghdad, Iraq
| |
Collapse
|
34
|
Davola ME, Mossman KL. Oncolytic viruses: how "lytic" must they be for therapeutic efficacy? Oncoimmunology 2019; 8:e1581528. [PMID: 31069150 PMCID: PMC6492965 DOI: 10.1080/2162402x.2019.1596006] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/22/2019] [Accepted: 03/07/2019] [Indexed: 12/21/2022] Open
Abstract
Oncolytic viruses (OVs) preferentially target and kill cancer cells without affecting healthy cells through a multi-modal mechanism of action. While historically the direct killing activity of OVs was considered the primary mode of action, initiation or augmentation of a host antitumor immune response is now considered an essential aspect of oncolytic virotherapy. To improve oncolytic virotherapy, many studies focus on increasing virus replication and spread. In this article, we open for discussion the traditional dogma that correlates replication with the efficacy of OVs, pointing out several examples that oppose this principle.
Collapse
Affiliation(s)
- Maria Eugenia Davola
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Michael DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Karen Louise Mossman
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Michael DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
35
|
Ma W, He H, Wang H. Oncolytic herpes simplex virus and immunotherapy. BMC Immunol 2018; 19:40. [PMID: 30563466 PMCID: PMC6299639 DOI: 10.1186/s12865-018-0281-9] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 12/06/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Oncolytic viruses have been proposed to be employed as a potential treatment of cancer. Well targeted, they will serve the purpose of cracking tumor cells without causing damage to normal cells. In this category of oncolytic viral drugs human pathogens herpes simplex virus (HSV) is especially suitable for the cause. Although most viral infection causes antiviral reaction in the host, HSV has multiple mechanisms to evade those responses. Powerful anti-tumor effect can thus be achieved via genetic manipulation of the HSV genes involved in this evading mechanism, namely deletions or mutations that adapt its function towards a tumor microenvironment. Currently, oncolytic HSV (oHSV) is widely use in clinical; moreover, there's hope that its curative effect will be further enhanced through the combination of oHSV with both traditional and emerging therapeutics. RESULTS In this review, we provide a summary of the HSV host antiviral response evasion mechanism, HSV expresses immune evasion genes such as ICP34.5, ICP0, Us3, which are involved in inducing and activating host responses, so that the virus can evade the immune system and establish effective long-term latent infection; we outlined details of the oHSV strains generated by removing genes critical to viral replication such as ICP34.5, ICP0, and inserting therapeutic genes such as LacZ, granulocyte macrophage colony-stimulating factor (GM-CSF); security and limitation of some oHSV such G207, 1716, OncoVEX, NV1020, HF10, G47 in clinical application; and the achievements of oHSV combined with immunotherapy and chemotherapy. CONCLUSION We reviewed the immunotherapy mechanism of the oHSV and provided a series of cases. We also pointed out that an in-depth study of the application of oHSV in cancer treatment will potentially benefits cancer patients more.
Collapse
Affiliation(s)
- Wenqing Ma
- Ruminant Diseases Research Center, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, 250014, China
| | - Hongbin He
- Ruminant Diseases Research Center, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, 250014, China.
| | - Hongmei Wang
- Ruminant Diseases Research Center, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, 250014, China.
| |
Collapse
|
36
|
Yang X, Huang B, Deng L, Hu Z. Progress in gene therapy using oncolytic vaccinia virus as vectors. J Cancer Res Clin Oncol 2018; 144:2433-2440. [PMID: 30293118 DOI: 10.1007/s00432-018-2762-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 09/28/2018] [Indexed: 01/06/2023]
Abstract
BACKGROUND Vaccinia virus was widely used in the World Health Organization's smallpox eradication campaign and is currently a promising vector for gene therapy owing to its unique characteristics. Vaccinia virus can selectively replicate and propagate productively in tumor cells, resulting in oncolysis. In addition, rapid viral particle production, wide host range, large genome size (approximately 200 kb), and safe handling render vaccinia virus a suitable vector for gene therapy. MATERIALS AND METHODS Cancer vaccines and gene therapy are being studied in clinical trials and experiment researches. However, we put forward unique challenges of optimal selection of foreign genes, administration and modification of VACV, personalized medicine, and other existing problems, based on current researches and our own experiments. CONCLUSION This review presents an overview of the vaccinia virus from its mechanisms to medical researches and clinical trials. We believe that the solution to these problems will contribute to understanding mechanisms of VACV and provide a theoretical basis for clinical treatment.
Collapse
Affiliation(s)
- Xue Yang
- Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Children's Hospital, Wuxi, 214023, Jiangsu, China
| | - Biao Huang
- School of Life Science, Zhejiang Sci-Tech University, Zhejiang, 310018, China
| | - Lili Deng
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, Jiangsu, China
| | - Zhigang Hu
- Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Children's Hospital, Wuxi, 214023, Jiangsu, China.
| |
Collapse
|
37
|
A Novel Chimeric Oncolytic Virus Vector for Improved Safety and Efficacy as a Platform for the Treatment of Hepatocellular Carcinoma. J Virol 2018; 92:JVI.01386-18. [PMID: 30232179 DOI: 10.1128/jvi.01386-18] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 09/12/2018] [Indexed: 11/20/2022] Open
Abstract
Oncolytic viruses represent an exciting new aspect of the evolving field of cancer immunotherapy. We have engineered a novel hybrid vector comprising vesicular stomatitis virus (VSV) and Newcastle disease virus (NDV), named recombinant VSV-NDV (rVSV-NDV), wherein the VSV backbone is conserved but its glycoprotein has been replaced by the hemagglutinin-neuraminidase (HN) and the modified, hyperfusogenic fusion (F) envelope proteins of recombinant NDV. In comparison to wild-type VSV, which kills cells through a classical cytopathic effect, the recombinant virus is able to induce tumor-specific syncytium formation, allowing efficient cell-to-cell spread of the virus and a rapid onset of immunogenic cell death. Furthermore, the glycoprotein exchange substantially abrogates the off-target effects in brain and liver tissue associated with wild-type VSV, resulting in a markedly enhanced safety profile, even in immune-deficient NOD.CB17-prkdcscid/NCrCrl (NOD-SCID) mice, which are highly susceptible to wild-type VSV. Although NDV causes severe pathogenicity in its natural avian hosts, the incorporation of the envelope proteins in the chimeric rVSV-NDV vector is avirulent in embryonated chicken eggs. Finally, systemic administration of rVSV-NDV in orthotopic hepatocellular carcinoma (HCC)-bearing immune-competent mice resulted in significant survival prolongation. This strategy, therefore, combines the beneficial properties of the rapidly replicating VSV platform with the highly efficient spread and immunogenic cell death of a fusogenic virus without risking the safety and environmental threats associated with either parental vector. Taking the data together, rVSV-NDV represents an attractive vector platform for clinical translation as a safe and effective oncolytic virus.IMPORTANCE The therapeutic efficacy of oncolytic viral therapy often comes as a tradeoff with safety, such that potent vectors are often associated with toxicity, while safer viruses tend to have attenuated therapeutic effects. Despite promising preclinical data, the development of VSV as a clinical agent has been substantially hampered by the fact that severe neurotoxicity and hepatotoxicity have been observed in rodents and nonhuman primates in response to treatment with wild-type VSV. Although NDV has been shown to have an attractive safety profile in humans and to have promising oncolytic effects, its further development has been severely restricted due to the environmental risks that it poses. The hybrid rVSV-NDV vector, therefore, represents an extremely promising vector platform in that it has been rationally designed to be safe, with respect to both the recipient and the environment, while being simultaneously effective, both through its direct oncolytic actions and through induction of immunogenic cell death.
Collapse
|
38
|
Yang M, Yang C, Tao Y, Tang J, Huang Q, Guo W, Feng S, Jiang A, Xu X, Jiang G, Liu Y. Combination therapy with F5/35 fiber chimeric conditionally replicative adenoviruses expressing IL-24 enhances the antitumor effect of temozolomide against melanoma. Cancer Med 2018; 7:5928-5942. [PMID: 30406970 PMCID: PMC6308089 DOI: 10.1002/cam4.1843] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 09/28/2018] [Accepted: 10/04/2018] [Indexed: 12/28/2022] Open
Abstract
Background Temozolomide (TMZ) is widely used to treat melanoma; however, response rates to TMZ are low because of rapid and frequent resistance. Conditionally, replicative adenoviruses (CRAds) are an effective and promising approach. The receptor for adenovirus is coxsackie‐adenovirus receptor (CAR), which is poorly expressed in most cells. However, CD46, which is the receptor of species B adenoviruses (Ads), is highly expressed in many cells. Methods We constructed CRAd F5/35‐ZD55‐IL‐24, which uses the viral receptors CAR and CD46 for entry into cells. We investigated the antitumor effect of F5/35‐ZD55‐IL‐24 in combination with TMZ to treat melanoma in vitro and in vivo. Results The \results indicated that F5/35‐ZD55‐IL‐24 in combination with TMZ produced additive or synergistic antitumor and pro‐apoptotic effects in melanoma cells. The combination of F5/35‐ZD55‐IL‐24 and TMZ significantly inhibited the growth of melanoma in vivo. In addition, the antitumor effect of F5/35‐ZD55‐IL‐24 was superior to that of ZD55‐IL‐24 and ZD55‐IL‐24 combined with TMZ. Conclusions The use of F5/35‐ZD55‐IL‐24 in conjunction with TMZ is a promising approach for anti‐melanoma therapy. Our results indicated that F5/35‐ZD55‐IL‐24 in combination with TMZ produced additive or synergistic antitumor effect and pro‐apoptotic effect in melanoma cells highly expressed CD46. The combination of F5/35‐ZD55‐IL‐24 and TMZ significantly inhibited the growth of melanoma in vivo. We also found the antitumor effect of F5/35‐ZD55‐IL‐24 was superior to ZD55‐IL‐24, the combination of F5/35‐ZD55‐IL‐24 and TMZ had a more significant antitumor effect than ZD55‐IL‐24 combining with TMZ.
Collapse
Affiliation(s)
- Ming Yang
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Department of Radiotherapy, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Chunsheng Yang
- Department of Dermatology, The Second People's Hospital of Huai'an, Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| | - Yingkai Tao
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jianqin Tang
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Qian Huang
- Department of Radiotherapy, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wenwen Guo
- Department of Radiotherapy, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Shouxin Feng
- Department of Radiotherapy, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Aijun Jiang
- Department of Radiotherapy, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xifeng Xu
- Department of Radiotherapy, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Guan Jiang
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yanqun Liu
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
39
|
Peng J, Wang S, Fan W, Li S, Wu Y, Mou X, Wang J, Tong X. Synergistic suppression effect on tumor growth of acute myeloid leukemia by combining cytarabine with an engineered oncolytic vaccinia virus. Onco Targets Ther 2018; 11:6887-6900. [PMID: 30410347 PMCID: PMC6199215 DOI: 10.2147/ott.s172037] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND In consideration of the drug resistance and side effects associated with cytarabine, one of the most effective drugs for the treatment of acute myeloid leukemia (AML), there is a need for safer and effective strategies. METHODS In the present investigation, we fabricated a new oncolytic vaccinia virus (oVV-ING4), which expresses the inhibitor of growth family member 4 (ING4) and explored its antitumor activity individually and in combination with cytarabine in AML cells. RESULTS The experiments confirmed that oVV can efficiently and specifically infect leukemia cells, and augment the ING4 gene expression. Flow cytometry and western blot demonstrated that oVV-ING4 enhances apoptosis and G2/M phase arrest in AML cells, and causes remarkable cancer cell death. In addition, the synergistic efficiency of oVV-ING4 and cytarabine was investigated in vitro and in vivo; the combination significantly inhibited the survival of leukemia cells in vitro and xenografted KG-1 AML tumor growth in vivo. CONCLUSION In brief, oVV-ING4 can increase the sensitivity of leukemia cells to cytarabine and induce cell apoptosis in vitro and in vivo. Thus, oVV-ING4 may be a promising therapeutic candidate for leukemia and in combination with cytarabine represents a potential antitumor therapy.
Collapse
Affiliation(s)
- Jiamin Peng
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China,
| | - Shibing Wang
- Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, China,
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou 310014, China,
| | - Weimin Fan
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China,
| | - Shuangshuang Li
- Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, China,
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou 310014, China,
| | - Yi Wu
- Department of Hematology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Xiaozhou Mou
- Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, China,
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou 310014, China,
| | - Jianchao Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China,
- Department of Clinical Laboratory, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310015, China
| | - Xiangmin Tong
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China,
- Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, China,
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou 310014, China,
| |
Collapse
|
40
|
Jung MY, Offord CP, Ennis MK, Kemler I, Neuhauser C, Dingli D. In Vivo Estimation of Oncolytic Virus Populations within Tumors. Cancer Res 2018; 78:5992-6000. [PMID: 30115692 DOI: 10.1158/0008-5472.can-18-0447] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/31/2018] [Accepted: 08/07/2018] [Indexed: 01/04/2023]
Abstract
The use of replication-competent viruses as oncolytic agents is rapidly expanding, with several oncolytic viruses approved for cancer therapy. As responses to therapy are highly variable, understanding the dynamics of therapy is critical for optimal application of virotherapy in practice. Although mathematical models have been developed to understand the dynamics of tumor virotherapy, a scarcity of in vivo data has made difficult parametrization of these models. To tackle this problem, we studied the in vitro and in vivo spread of two oncolytic measles viruses that induce expression of the sodium iodide symporter (NIS) in cells. NIS expression enabled infected cells to concentrate radioactive isotopes that could be reproducibly and quantitatively imaged using SPECT/CT. We observed a strong linear relationship in vitro between infectious virus particles, viral N and NIS gene expression, and radioactive isotope uptake. In vivo radioisotope uptake was highly correlated with viral N and NIS gene expression. Similar expression patterns between viral N and NIS gene expression in vitro and in vivo implied that the oncolytic virus behaved similarly in both scenarios. Significant titers of viable virus were consistently isolated from tumors explanted from mice that had been injected with oncolytic measle viruses. We observed a weaker but positive in vivo relationship between radioisotope uptake and the viable virus titer recovered from tumors; this was likely due to anisotropies in the viral distribution in vivo These data suggest that methods that enable quantitation of in vivo anisotropies are required for continuing development of oncolytic virotherapy.Significance: These findings address a fundamental gap in our knowledge of oncolytic virotherapy by presenting technology that gives insight into the behavior of oncolytic viruses in vivo Cancer Res; 78(20); 5992-6000. ©2018 AACR.
Collapse
Affiliation(s)
- Mi-Yeon Jung
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Chetan P Offord
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Matthew K Ennis
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Iris Kemler
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Claudia Neuhauser
- College of Biological Sciences, University of Minnesota, Twin Cities, Minnesota.,Bioinformatics and Computational Biology Program, University of Minnesota Rochester, Rochester, Minnesota
| | - David Dingli
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota. .,Bioinformatics and Computational Biology Program, University of Minnesota Rochester, Rochester, Minnesota.,Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
41
|
Cho E, Ryu EJ, Jiang F, Jeon UB, Cho M, Kim CH, Kim M, Kim ND, Hwang TH. Preclinical safety evaluation of hepatic arterial infusion of oncolytic poxvirus. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:2467-2474. [PMID: 30122903 PMCID: PMC6087018 DOI: 10.2147/dddt.s171269] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Purpose Oncolytic poxvirus has shown promise in treating various solid tumors, such as liver cancer, and administration of oncolytic poxvirus via the hepatic artery may provide more survival benefits than other routes of administration. However, there is a lack of safety information to guide the application of hepatic arterial infusion (HAI) of oncolytic poxvirus in human studies. To investigate the acute and chronic toxicity of HAI administration of oncolytic poxvirus in animals and provide safety information for future human studies. Methods VVtk-, a vaccinia poxvirus with inactivated thymidine kinase gene, was administered via HAI to rabbits with normal liver function under angiography (1×108 or 1×109 pfu), and rats with N-nitrosomorpholine-induced precancerous liver cirrhosis under open surgery (1×108 pfu). Body weights and survival were monitored and blood samples were collected for hematological and biochemical tests. Distribution of A56 (a specific marker for poxvirus infection) in rabbit organs was evaluated using immunofluorescence assays. Results HAI of high doses of VVtk- did not cause any acute or chronic changes in body weight, survival or in biochemical, hematological tests in the 2 animal models, and none of the changes showed dose dependency (in rabbit study), or were influenced by liver cirrhosis (in rat study). A56 was not detected in any of the major rabbit organs. Conclusion HAI may provide a safe alternative route of oncolytic poxvirus administration for human studies.
Collapse
Affiliation(s)
- Euna Cho
- Department of Pharmacology and Medical Research Center (MRC), Pusan National University School of Medicine, Yangsan, Korea, .,Department of Research and Development, Bionoxx Inc, Seongnam-si, Korea,
| | - Eun Jin Ryu
- Department of Research and Development, Bionoxx Inc, Seongnam-si, Korea, .,Department of Radiology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Fen Jiang
- Department of Research and Development, Bionoxx Inc, Seongnam-si, Korea, .,School of Pharmaceutical Science (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Ung Bae Jeon
- Department of Radiology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Mong Cho
- Department of Pharmacology and Medical Research Center (MRC), Pusan National University School of Medicine, Yangsan, Korea, .,Department of Research and Development, Bionoxx Inc, Seongnam-si, Korea,
| | - Cy Hyun Kim
- Department of Pharmacology and Medical Research Center (MRC), Pusan National University School of Medicine, Yangsan, Korea,
| | - Miyoung Kim
- Department of Pharmacology and Medical Research Center (MRC), Pusan National University School of Medicine, Yangsan, Korea,
| | - Nam Deuk Kim
- Department of Pharmacy and Pusan Cancer Research Center, Pusan National University, Busan, Korea
| | - Tae-Ho Hwang
- Department of Pharmacology and Medical Research Center (MRC), Pusan National University School of Medicine, Yangsan, Korea, .,Department of Research and Development, Bionoxx Inc, Seongnam-si, Korea,
| |
Collapse
|
42
|
Chen CY, Hutzen B, Wedekind MF, Cripe TP. Oncolytic virus and PD-1/PD-L1 blockade combination therapy. Oncolytic Virother 2018; 7:65-77. [PMID: 30105219 PMCID: PMC6074764 DOI: 10.2147/ov.s145532] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Oncolytic viruses are lytic for many types of cancers but are attenuated or replication-defective in normal tissues. Aside from tumor lysis, oncolytic viruses can induce host immune responses against cancer cells and may thus be viewed as a form of immunotherapy. Although recent successes with checkpoint inhibitors have shown that enhancing antitumor immunity can be effective, the dynamic nature of the immunosuppressive tumor microenvironment presents significant hurdles to the broader application of these therapies. Targeting one immune-suppressive pathway may not be sufficient to eliminate tumors. Here we focus on the development of the combination of oncolytic virotherapy with checkpoint inhibitors designed to target the programmed cell death protein 1 and programmed cell death ligand 1 signaling axis. We also discuss future directions for the clinical application of this novel combination therapy.
Collapse
Affiliation(s)
- Chun-Yu Chen
- Department of Pediatrics, Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital,
| | - Brian Hutzen
- Department of Pediatrics, Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital,
| | - Mary F Wedekind
- Department of Pediatrics, Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, .,Division of Hematology/Oncology/Blood and Marrow Transplantation, Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA,
| | - Timothy P Cripe
- Department of Pediatrics, Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, .,Division of Hematology/Oncology/Blood and Marrow Transplantation, Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA,
| |
Collapse
|
43
|
Deng L, Fan J, Ding Y, Zhang J, Zhou B, Zhang Y, Huang B. Oncolytic efficacy of thymidine kinase-deleted vaccinia virus strain Guang9. Oncotarget 2018; 8:40533-40543. [PMID: 28465492 PMCID: PMC5522336 DOI: 10.18632/oncotarget.17125] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 04/04/2017] [Indexed: 11/29/2022] Open
Abstract
Oncolytic virotherapy is being developed as a promising platform for cancer therapy due to its ability to lyse cancer cells in a tumor-specific manner. Vaccinia virus has been used as a live vaccine in the smallpox eradication program and now is being potential in cancer therapy with a great safety profile. Vaccinia strain Guang9 (VG9) is an attenuated Chinese vaccinia virus and its oncolytic efficacy has been evaluated in our previous study. To improve the tumor selectivity and oncolytic efficacy, we here developed a thymidine kinase (TK)-deleted vaccinia virus based on Guang9 strain. The viral replication, marker gene expression and cytotoxicity in various cell lines were evaluated; antitumor effects in vivo were assessed in multiple tumor models. In vitro, the TK-deleted vaccinia virus replicated rapidly, but the cytotoxicity varied in different cell lines. It was notably attenuated in normal cells and resting cells in vitro, while tumor-selectively replicated in vivo. Significant antitumor effects were observed both in murine melanoma tumor model and human hepatoma tumor model. It significantly inhibited the growth of subcutaneously implanted tumors and prolonged the survival of tumor-bearing mice. Collectively, TK-deleted vaccinia strain Guang9 is a promising constructive virus vector for tumor-directed gene therapy and will be a potential therapeutic strategy in cancer treatment.
Collapse
Affiliation(s)
- Lili Deng
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Jun Fan
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Yuedi Ding
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Jue Zhang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Bin Zhou
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Yi Zhang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Biao Huang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| |
Collapse
|
44
|
Vile RG. The Immune System in Oncolytic Immunovirotherapy: Gospel, Schism and Heresy. Mol Ther 2018; 26:942-946. [PMID: 29573976 DOI: 10.1016/j.ymthe.2018.03.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Richard G Vile
- Department of Molecular Medicine and Immunology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
45
|
Oncotargeting by Vesicular Stomatitis Virus (VSV): Advances in Cancer Therapy. Viruses 2018; 10:v10020090. [PMID: 29473868 PMCID: PMC5850397 DOI: 10.3390/v10020090] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 02/12/2018] [Accepted: 02/15/2018] [Indexed: 12/28/2022] Open
Abstract
Modern oncotherapy approaches are based on inducing controlled apoptosis in tumor cells. Although a number of apoptosis-induction approaches are available, site-specific delivery of therapeutic agents still remain the biggest hurdle in achieving the desired cancer treatment benefit. Additionally, systemic treatment-induced toxicity remains a major limiting factor in chemotherapy. To specifically address drug-accessibility and chemotherapy side effects, oncolytic virotherapy (OV) has emerged as a novel cancer treatment alternative. In OV, recombinant viruses with higher replication capacity and stronger lytic properties are being considered for tumor cell-targeting and subsequent cell lysing. Successful application of OVs lies in achieving strict tumor-specific tropism called oncotropism, which is contingent upon the biophysical interactions of tumor cell surface receptors with viral receptors and subsequent replication of oncolytic viruses in cancer cells. In this direction, few viral vector platforms have been developed and some of these have entered pre-clinical/clinical trials. Among these, the Vesicular stomatitis virus (VSV)-based platform shows high promise, as it is not pathogenic to humans. Further, modern molecular biology techniques such as reverse genetics tools have favorably advanced this field by creating efficient recombinant VSVs for OV; some have entered into clinical trials. In this review, we discuss the current status of VSV based oncotherapy, challenges, and future perspectives regarding its therapeutic applications in the cancer treatment.
Collapse
|
46
|
Cockle JV, Scott KJ. What is oncolytic virotherapy? Arch Dis Child Educ Pract Ed 2018; 103:43-45. [PMID: 28588023 DOI: 10.1136/archdischild-2016-311922] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 04/20/2017] [Indexed: 01/29/2023]
Affiliation(s)
- Julia V Cockle
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK.,Yorkshire Regional Centre for Paediatric Oncology and Haematology, Leeds General Infirmary, Leeds, UK
| | - Karen J Scott
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| |
Collapse
|
47
|
A hypoxia- and telomerase-responsive oncolytic adenovirus expressing secretable trimeric TRAIL triggers tumour-specific apoptosis and promotes viral dispersion in TRAIL-resistant glioblastoma. Sci Rep 2018; 8:1420. [PMID: 29362367 PMCID: PMC5780382 DOI: 10.1038/s41598-018-19300-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 12/27/2017] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma is a highly aggressive and malignant type of cancer that is apoptosis resistant and difficult to cure by conventional cancer therapies. In this regard, an oncolytic adenovirus that selectively targets the tumour tissue and induces tumour cell lysis is a promising treatment option. We designed and constructed a hypoxia-responsive and cancer-specific modified human telomerase reverse transcriptase (H5CmTERT) promoter to drive replication of an oncolytic adenovirus (H5CmTERT-Ad). To enhance the anti-tumour efficacy of H5CmTERT-Ad against malignant glioblastoma, we also generated an H5CmTERT-Ad expressing secretable trimeric tumour necrosis factor-related apoptosis-inducing ligand (H5CmTERT-Ad/TRAIL). H5CmTERT promoter-regulated oncolytic adenoviruses showed cancer-specific and superior cell-killing effect in contrast to a cognate control oncolytic adenovirus replicating under the control of the endogenous adenovirus promoter. The cancer cell-killing effects of H5CmTERT-Ad and H5CmTERT-Ad/TRAIL were markedly higher during hypoxia than normoxia owing to hypoxia responsiveness of the promoter. H5CmTERT-Ad/TRAIL showed more potent anti-tumour efficacy than H5CmTERT-Ad did in a xenograft model of TRAIL-resistant subcutaneous and orthotopic glioblastoma through superior induction of apoptosis and more extensive virus distribution in the tumour tissue. Altogether, our findings show that H5CmTERT-Ad/TRAIL can promote dispersion of an oncolytic adenovirus through robust induction of apoptosis in a highly TRAIL-resistant glioblastoma.
Collapse
|
48
|
Oncolytic Viruses: T-VEC and Others. Oncoimmunology 2018. [DOI: 10.1007/978-3-319-62431-0_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
49
|
Yang M, Yang CS, Guo W, Tang J, Huang Q, Feng S, Jiang A, Xu X, Jiang G, Liu YQ. A novel fiber chimeric conditionally replicative adenovirus-Ad5/F35 for tumor therapy. Cancer Biol Ther 2017; 18:833-840. [PMID: 29144842 DOI: 10.1080/15384047.2017.1395115] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Significant progress has been made in the diagnosis and treatment of cancer; however, significant challenges remain. Conditionally replicating adenoviruses (CRAds), which not only kill cancer cells, but also serve as vectors to express therapeutic genes, are a novel and effective method to treat cancer. However, most adenoviruses are Ad5, which infect cells through the coxsackie and adenovirus receptor (CAR). The transduction efficacy of Ad5 is restricted because of the absent or low expression of CAR on several cancer cells. Ad serotype 35 has a different tropism pattern to Ad5. Ad35 attaches to cells via a non-CAR receptor, CD46, which is expressed widely on most tumor cells. Thus, chimeric adenoviral vectors consisting of the knob and shaft of Ad35 combined with Ad5 have been constructed. The chimeric fiber adenoviral vectors can transduce CAR-positive and CAR-negative cell lines. In this review, we explore the application of the novel fiber chimeric conditionally replicative adenovirus-Ad5/F35 in tumor therapy in terms of safety, mechanism, transduction efficacy, and antitumor effect.
Collapse
Affiliation(s)
- Ming Yang
- a Department of Radiotherapy , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China.,b Department of Oncology , Affiliated Nanyang Second General Hospital , Nanyang , China
| | - Chun Sheng Yang
- c Department of Dermatology , Affiliated Huai'an Hospital of Xuzhou Medical University , the Second People's Hospital of Huai'an, Huai'an , China
| | - WenWen Guo
- a Department of Radiotherapy , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China
| | - JianQin Tang
- d Department of Dermatology , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China
| | - Qian Huang
- a Department of Radiotherapy , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China
| | - ShouXin Feng
- a Department of Radiotherapy , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China
| | - AiJun Jiang
- a Department of Radiotherapy , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China
| | - XiFeng Xu
- a Department of Radiotherapy , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China
| | - Guan Jiang
- d Department of Dermatology , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China
| | - Yan Qun Liu
- d Department of Dermatology , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China
| |
Collapse
|
50
|
Yoon AR, Hong J, Yun CO. Adenovirus-mediated decorin expression induces cancer cell death through activation of p53 and mitochondrial apoptosis. Oncotarget 2017; 8:76666-76685. [PMID: 29100340 PMCID: PMC5652734 DOI: 10.18632/oncotarget.20800] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 08/23/2017] [Indexed: 12/15/2022] Open
Abstract
Decorin (DCN) is a small leucine-rich proteoglycan that plays an important role in the regulation of apoptosis, proliferation, intercellular contact, and cell migration. Here we have investigated the detailed mechanism of apoptotic cell death induced by DCN expression. A marked increase in cytotoxicity was observed for both DCN-expressing replication-incompetent (dE1/DCN) and -competent (dB/DCN) adenoviruses (Ads) compared to the corresponding control Ads. FACS and TUNEL assays revealed that the expression of DCN induced apoptotic cell death. Specifically, the expression and stability of p53 were increased by DCN. In addition, western blot data showed that DCN expression activated mitochondrial apoptosis by increasing the expression level of p53. Similarly, DCN-expressing oncolytic Ads induced a greater antitumor effect in a murine xenograft model compared with control Ads. Tissue staining and western blot data from in vivo experiments demonstrated significantly higher levels of apoptosis in tumor tissues from mice treated with DCN-expressing Ads compared to those treated with control Ads. Collectively, these data support that cell killing effect is enhanced with Ad-mediated DCN expression via the induction of p53-mediated mitochondrial apoptosis, which could be a valuable benefit for antitumor efficacy.
Collapse
Affiliation(s)
- A-Rum Yoon
- Department of Bioengineering, College of Engineering, Hanyang University, Seongdong-gu, Seoul 04763, Korea
| | - JinWoo Hong
- Department of Bioengineering, College of Engineering, Hanyang University, Seongdong-gu, Seoul 04763, Korea
| | - Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University, Seongdong-gu, Seoul 04763, Korea
| |
Collapse
|