1
|
Gao Y, Zou Y, Wu C, Tao J, Nie Z, Yan J, Wang P, Huang X. Comparative evaluation of immunomodulatory cytokines for oncolytic therapy based on a high-efficient platform for oHSV1 reconstruction. Virol J 2025; 22:133. [PMID: 40325455 PMCID: PMC12054163 DOI: 10.1186/s12985-025-02758-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 04/22/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) presents significant therapeutic challenges due to its immunosuppressive tumor microenvironment (TME). Oncolytic herpes simplex virus type 1 (oHSV1) offers dual mechanisms of tumor lysis and immune activation, yet the optimal cytokine payloads for TNBC remain undefined. METHODS We developed a CRISPR/Cas9-mediated platform for high-efficiency oHSV1 engineering, replacing the ICP47 locus with murine IFN-γ, GM-CSF, or IL-15Rα/IL-15 fusion protein (IL15Fu). Constructs were validated for cytokine secretion, MHC modulation, and cytotoxicity in 4T1 TNBC and a panel of human cancer cell lines. Antitumor efficacy and immune remodeling were evaluated in a syngeneic 4T1 model using RNA sequencing and flow cytometry. RESULTS The CRISPR platform achieved 62.5-71.4% homologous recombination efficiency, enabling rapid virus construction. In vitro, OV-IFNG exhibited upregulated MHC I/II expression and potent cytotoxicity, while OV-GMCSF attenuated oncolysis in subsets of breast cancer cell lines. In the 4T1 model, OV-IL15Fu modestly improved tumor control and extended survival without apparent toxicity, while OV-IFNG induced early mortality associated with systemic toxicity. Transcriptomic profiling revealed divergent immune modulation: OV-IL15Fu enriched T cell/NK cytotoxicity pathways, OV-IFNG amplified cytokine/chemokine signaling, and OV-GMCSF paradoxically enhanced myeloid recruitment while inhibiting MHC-II pathways. Flow cytometry confirmed functional differences in immune activation: OV-IL15Fu expanding cytotoxic lymphocytes (CD8⁺ T/NK cells), OV-IFNG preferentially promote Th1 polarization and innate immune activation, and OV-GMCSF failed to activate T cells despite myeloid infiltration. CONCLUSIONS Our findings underscore the need for rational cytokine selection in oHSV1-based immunotherapy. While IFN-γ increased immunogenic markers, its systemic toxicity and myeloid effects may limit benefit. GM-CSF exacerbated immune suppression in this context, whereas IL15Fu showed favorable immunostimulatory properties without detectable toxicity. These data support IL15Fu as a contextually promising payload for further evaluation in TNBC-targeted oncolytic virotherapy.
Collapse
Affiliation(s)
- Yingzheng Gao
- Kunming Medical University, Kunming, Yunnan, China
- Key Laboratory of The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yufang Zou
- Kunming Medical University, Kunming, Yunnan, China
- Key Laboratory of The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Changjing Wu
- Kunming Medical University, Kunming, Yunnan, China
- Key Laboratory of The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Juan Tao
- Kunming Medical University, Kunming, Yunnan, China
- Key Laboratory of The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zuqing Nie
- Kunming Medical University, Kunming, Yunnan, China
- Key Laboratory of The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Jinyuan Yan
- Kunming Medical University, Kunming, Yunnan, China
- Key Laboratory of The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Pengfei Wang
- Kunming Medical University, Kunming, Yunnan, China.
- Key Laboratory of The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| | - Xinwei Huang
- Kunming Medical University, Kunming, Yunnan, China.
- Key Laboratory of The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| |
Collapse
|
2
|
Zhang J, Xiao Y, Zhang J, Yang Y, Zhang L, Liang F. Recent advances of engineered oncolytic viruses-based combination therapy for liver cancer. J Transl Med 2024; 22:3. [PMID: 38167076 PMCID: PMC10763442 DOI: 10.1186/s12967-023-04817-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024] Open
Abstract
Liver cancer is a major malignant tumor, which seriously threatens human health and increases the economic burden on patients. At present, gene therapy has been comprehensively studied as an excellent therapeutic measure in liver cancer treatment. Oncolytic virus (OV) is a kind of virus that can specifically infect and kill tumor cells. After being modified by genetic engineering, the specificity of OV infection to tumor cells is increased, and its influence on normal cells is reduced. To date, OV has shown its effectiveness and safety in experimental and clinical studies on a variety of tumors. Thus, this review primarily introduces the current status of different genetically engineered OVs used in gene therapy for liver cancer, focuses on the application of OVs and different target genes for current liver cancer therapy, and identifies the problems encountered in OVs-based combination therapy and the corresponding solutions, which will provide new insights into the treatment of liver cancer.
Collapse
Affiliation(s)
- Junhe Zhang
- Institutes of Health Central Plains, Xinxiang Medical University, No. 601 Jinsui Road, Xinxiang, 453003, Henan Province, China.
- Henan Key Laboratory of Neurorestoratology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, China.
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China.
| | - Yunxi Xiao
- Institutes of Health Central Plains, Xinxiang Medical University, No. 601 Jinsui Road, Xinxiang, 453003, Henan Province, China
| | - Jie Zhang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Yun Yang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Liao Zhang
- Institutes of Health Central Plains, Xinxiang Medical University, No. 601 Jinsui Road, Xinxiang, 453003, Henan Province, China
| | - Fan Liang
- Institutes of Health Central Plains, Xinxiang Medical University, No. 601 Jinsui Road, Xinxiang, 453003, Henan Province, China
| |
Collapse
|
3
|
Khalaf AT, Wan J, Wei H, Fubing S, Zainol J, Kadir SYA, Liang M. Vector-Mediated Cancer Gene Therapy Reduces Toxicity and Inhibition of Lung Carcinoma Growth in Nude Mice. Appl Biochem Biotechnol 2024; 196:261-274. [PMID: 37119504 DOI: 10.1007/s12010-023-04463-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2023] [Indexed: 05/01/2023]
Abstract
Replication-competent oncolytic adenovirus (TOA2) gene therapy is a recently introduced anti-tumor treatment regimen with superior results. The biodistribution studies of virus vector-based medicine seem more cautious and have been given much attention recently in terms of its quality and safety in preclinical trials. The current study determined the biodistribution and safety of a replication-competent adenovirus in different organs to predict its toxicity threshold. The present study has used TOA2, while biodistribution analysis was performed in human lung carcinoma A549-induced tumor-bearing nude mice model. Intratumoral injection was applied onto tumor-bearing mice with the adenovirus (3×1010 VP per mouse). Mice were sacrificed at the end of the experiment and the organs were dissected. Biodistribution analysis was done with complete hexon gene detection in each organ using quantitative real-time polymerase chain reaction (qRT-PCR). The biodistribution and concentration profiles showed that the TOA2 is well distributed in the entire tumor tissue. After dose 3 at day 11, the concentration of the virus has increased in the tumor tissue from 2240.54 (± 01.69) copies/100 ng genome to 13,120.28 (± 88.21) copies/100 ng genome on the 18th day, which eventually approached 336.45 (± 23.41) copies/100ng genome on the day 36. On the contrary, the concentration of the same decreased in the order of the liver, kidney, spleen, lung, and heart over time but no distributional traces in gonads. But the concentration found decreased dramatically in blood and other organs, while at the end of the experiment no detectable distribution was seen besides tumor tissue. The study confirms that adenovirus-based tumor therapy using conditionally replicating competent oncolytic TOA2 exhibited great efficiency with no toxicity at all.
Collapse
Affiliation(s)
- Ahmad Taha Khalaf
- Basic Medical College, Chengdu University, Chengdu, 610106, Sichuan Province, China
| | - Jun Wan
- Affiliated Hospital & Clinical Medical College of Chengdu University, Chengdu, 610081, China
- Key Laboratory of Pattern Recognition and Intelligent Information Processing, Institutions of Higher Education of Sichuan Province, Chengdu University, Sichuan, 610106, Chengdu, China
| | - Hu Wei
- Hospital of Wuhan University, Wuhan, Hubei province, China
| | - Shen Fubing
- Basic Medical College, Chengdu University, Chengdu, 610106, Sichuan Province, China
| | | | | | - Min Liang
- Department of Thoracic Surgery, West China Fourth Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
4
|
Liu D, Che X, Wang X, Ma C, Wu G. Tumor Vaccines: Unleashing the Power of the Immune System to Fight Cancer. Pharmaceuticals (Basel) 2023; 16:1384. [PMID: 37895855 PMCID: PMC10610367 DOI: 10.3390/ph16101384] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
This comprehensive review delves into the rapidly evolving arena of cancer vaccines. Initially, we examine the intricate constitution of the tumor microenvironment (TME), a dynamic factor that significantly influences tumor heterogeneity. Current research trends focusing on harnessing the TME for effective tumor vaccine treatments are also discussed. We then provide a detailed overview of the current state of research concerning tumor immunity and the mechanisms of tumor vaccines, describing the complex immunological processes involved. Furthermore, we conduct an exhaustive analysis of the contemporary research landscape of tumor vaccines, with a particular focus on peptide vaccines, DNA/RNA-based vaccines, viral-vector-based vaccines, dendritic-cell-based vaccines, and whole-cell-based vaccines. We analyze and summarize these categories of tumor vaccines, highlighting their individual advantages, limitations, and the factors influencing their effectiveness. In our survey of each category, we summarize commonly used tumor vaccines, aiming to provide readers with a more comprehensive understanding of the current state of tumor vaccine research. We then delve into an innovative strategy combining cancer vaccines with other therapies. By studying the effects of combining tumor vaccines with immune checkpoint inhibitors, radiotherapy, chemotherapy, targeted therapy, and oncolytic virotherapy, we establish that this approach can enhance overall treatment efficacy and offset the limitations of single-treatment approaches, offering patients more effective treatment options. Following this, we undertake a meticulous analysis of the entire process of personalized cancer vaccines, elucidating the intricate process from design, through research and production, to clinical application, thus helping readers gain a thorough understanding of its complexities. In conclusion, our exploration of tumor vaccines in this review aims to highlight their promising potential in cancer treatment. As research in this field continues to evolve, it undeniably holds immense promise for improving cancer patient outcomes.
Collapse
Affiliation(s)
- Dequan Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (D.L.); (X.C.)
| | - Xiangyu Che
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (D.L.); (X.C.)
| | - Xiaoxi Wang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China;
| | - Chuanyu Ma
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (D.L.); (X.C.)
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (D.L.); (X.C.)
| |
Collapse
|
5
|
Immunotherapy by mesenchymal stromal cell delivery of oncolytic viruses for treating metastatic tumors. Mol Ther Oncolytics 2022; 25:78-97. [PMID: 35434272 PMCID: PMC8989711 DOI: 10.1016/j.omto.2022.03.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Oncolytic viruses (OVs) have emerged as a very promising anti-cancer therapeutic strategy in the past decades. However, despite their pre-clinical promise, many OV clinical evaluations for cancer therapy have highlighted the continued need for their improved delivery and targeting. Mesenchymal stromal cells (MSCs) have emerged as excellent candidate vehicles for the delivery of OVs due to their tumor-homing properties and low immunogenicity. MSCs can enhance OV delivery by protecting viruses from rapid clearance following administration and also by more efficiently targeting tumor sites, consequently augmenting the therapeutic potential of OVs. MSCs can function as “biological factories,” enabling OV amplification within these cells to promote tumor lysis following MSC-OV arrival at the tumor site. MSC-OVs can promote enhanced safety profiles and therapeutic effects relative to OVs alone. In this review we explore the general characteristics of MSCs as delivery tools for cancer therapeutic agents. Furthermore, we discuss the potential of OVs as immune therapeutics and highlight some of the promising applications stemming from combining MSCs to achieve enhanced delivery and anti-tumor effectiveness of OVs at different pre-clinical and clinical stages. We further provide potential pitfalls of the MSC-OV platform and the strategies under development for enhancing the efficacy of these emerging therapeutics.
Collapse
|
6
|
Le TMD, Yoon AR, Thambi T, Yun CO. Polymeric Systems for Cancer Immunotherapy: A Review. Front Immunol 2022; 13:826876. [PMID: 35273607 PMCID: PMC8902250 DOI: 10.3389/fimmu.2022.826876] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 01/24/2022] [Indexed: 12/13/2022] Open
Abstract
Immunotherapy holds enormous promise to create a new outlook of cancer therapy by eliminating tumors via activation of the immune system. In immunotherapy, polymeric systems play a significant role in improving antitumor efficacy and safety profile. Polymeric systems possess many favorable properties, including magnificent biocompatibility and biodegradability, structural and component diversity, easy and controllable fabrication, and high loading capacity for immune-related substances. These properties allow polymeric systems to perform multiple functions in immunotherapy, such as immune stimulants, modifying and activating T cells, delivery system for immune cargos, or as an artificial antigen-presenting cell. Among diverse immunotherapies, immune checkpoint inhibitors, chimeric antigen receptor (CAR) T cell, and oncolytic virus recently have been dramatically investigated for their remarkable success in clinical trials. In this report, we review the monotherapy status of immune checkpoint inhibitors, CAR-T cell, and oncolytic virus, and their current combination strategies with diverse polymeric systems.
Collapse
Affiliation(s)
- Thai Minh Duy Le
- Department of Bioengineering, College of Engineering, Hanayang University, Seoul, South Korea
| | - A-Rum Yoon
- Department of Bioengineering, College of Engineering, Hanayang University, Seoul, South Korea.,Institute of Nano Science and Technology (INST), Hanayang University, Seoul, South Korea.,Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul, South Korea
| | - Thavasyappan Thambi
- Department of Bioengineering, College of Engineering, Hanayang University, Seoul, South Korea
| | - Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanayang University, Seoul, South Korea.,Institute of Nano Science and Technology (INST), Hanayang University, Seoul, South Korea.,Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul, South Korea.,GeneMedicine CO., Ltd., Seoul, South Korea
| |
Collapse
|
7
|
Tian Y, Xie D, Yang L. Engineering strategies to enhance oncolytic viruses in cancer immunotherapy. Signal Transduct Target Ther 2022; 7:117. [PMID: 35387984 PMCID: PMC8987060 DOI: 10.1038/s41392-022-00951-x] [Citation(s) in RCA: 123] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 02/07/2023] Open
Abstract
Oncolytic viruses (OVs) are emerging as potentially useful platforms in treatment methods for patients with tumors. They preferentially target and kill tumor cells, leaving healthy cells unharmed. In addition to direct oncolysis, the essential and attractive aspect of oncolytic virotherapy is based on the intrinsic induction of both innate and adaptive immune responses. To further augment this efficacious response, OVs have been genetically engineered to express immune regulators that enhance or restore antitumor immunity. Recently, combinations of OVs with other immunotherapies, such as immune checkpoint inhibitors (ICIs), chimeric antigen receptors (CARs), antigen-specific T-cell receptors (TCRs) and autologous tumor-infiltrating lymphocytes (TILs), have led to promising progress in cancer treatment. This review summarizes the intrinsic mechanisms of OVs, describes the optimization strategies for using armed OVs to enhance the effects of antitumor immunity and highlights rational combinations of OVs with other immunotherapies in recent preclinical and clinical studies.
Collapse
Affiliation(s)
- Yaomei Tian
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, No. 17, Section 3, South Renmin Road, 610041, Chengdu, Sichuan, People's Republic of China
- College of Bioengineering, Sichuan University of Science & Engineering, No. 519, Huixing Road, 643000, Zigong, Sichuan, People's Republic of China
| | - Daoyuan Xie
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, No. 17, Section 3, South Renmin Road, 610041, Chengdu, Sichuan, People's Republic of China
| | - Li Yang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, No. 17, Section 3, South Renmin Road, 610041, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
8
|
He J, An Y, Qi J, Cui L, Yang K, Liu M, Qu B, Yan S, Yin J, Jing X, Dong H, Yu Q, Li D, Wu Y. The recombinant Newcastle disease virus Anhinga strain expressing human TRAIL exhibit antitumor effects on a glioma nude mice model. J Med Virol 2021; 93:3890-3898. [PMID: 32779745 DOI: 10.1002/jmv.26419] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 07/22/2020] [Accepted: 08/04/2020] [Indexed: 12/13/2022]
Abstract
Oncolytic virus therapy is perhaps the next major breakthrough in cancer treatment following the success in immunotherapy using immune checkpoint inhibitors. However, the potential oncolytic ability of the recombinant newcastle disease virus (NDV) Anhinga strain carried with tumor necrosis factor-related apoptosis inducing ligand (TRAIL) has not been fully explored at present. In the present study, the recombinant NDV/Anh-TRAIL that secretes soluble TRAIL was constructed and the experiment results suggested NDV/Anh-TRAIL as a promising candidate for glioma therapy. Growth kinetic and TRAIL secreted quantity of recombinant NDV/Anh-TRAIL virus were measured. Cytotoxic and cell apoptosis were analyzed for its anti-glioma therapy in vitro. Nude mice were used for the in vivo evaluation. Both tumor volume and mice behavior after injection were observed. The recombinant virus replicated with the same kinetics as the parental virus and the highest expression of TRAIL (77.8 ng/L) was found at 48 hours. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, a tetrazole and flow cytometry data revealed that the recombinant NDV/Anh-TRAIL (56.1 ± 8.2%) virus could induce a more severe apoptosis rate, when compared with the NDV wild type (37.2 ± 7.0%) and mock (7.0 ± 1.8%) groups (P < .01), in U251 cells. Furthermore, in the present animal study, the average tumor volume was smaller in the NDV/Anh-TRAIL group (97.21 mm3 ), when compared with the NDV wild type (205.03 mm3 , P < .05) and PBS (310.30 mm3 , P < .01) groups.
Collapse
Affiliation(s)
- Jinjiao He
- College of Life Science, Northeast Agricultural University, Harbin, Heilongjiang, China
- School of Life sciences & Basic Medicine, Xinxiang University, Xinxiang, Henan, China
| | - Ying An
- Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianying Qi
- College of Life Sciences, Henan Normal University, Xinxiang, Henan, China
| | - Lin Cui
- College of Life Science, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Kai Yang
- Knowledge Management Center Nutrition & Health Research Institute, COFCO Corporation, Beijing, China
| | - Mingyao Liu
- College of Life Science, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Bo Qu
- College of Life Science, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Shijun Yan
- College of Life Science, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Jiechao Yin
- College of Life Science, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Xiaohui Jing
- College of Life Science, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Hui Dong
- College of Life Science, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Qingzhong Yu
- Southeast Poultry Research Laboratory, Agricultural Research Service, United States Department of Agriculture, Athens, Georgia
| | - Deshan Li
- College of Life Science, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Yunzhou Wu
- College of Life Science, Northeast Agricultural University, Harbin, Heilongjiang, China
| |
Collapse
|
9
|
Abstract
Tumors represent a hostile environment for the effector cells of cancer immunosurveillance. Immunosuppressive receptors and soluble or membrane-bound ligands are abundantly exposed and released by malignant entities and their stromal accomplices. As a consequence, executioners of antitumor immunity inefficiently navigate across cancer tissues and fail to eliminate malignant targets. By inducing immunogenic cancer cell death, oncolytic viruses profoundly reshape the tumor microenvironment. They trigger the local spread of danger signals and tumor-associated (as well as viral) antigens, thus attracting antigen-presenting cells, promoting the activation and expansion of lymphocytic populations, facilitating their infiltration in the tumor bed, and reinvigorating cytotoxic immune activity. The present review recapitulates key chemokines, growth factors and other cytokines that orchestrate this ballet of antitumoral leukocytes upon oncolytic virotherapy.
Collapse
Affiliation(s)
- Jonathan G Pol
- Centre de Recherche des Cordeliers, Equipe 11 labellisée par la Ligue Nationale contre le Cancer, INSERM, Sorbonne Université, Université de Paris, Paris, France; Gustave Roussy Cancer Campus, Metabolomics and Cell Biology Platforms, Villejuif, France.
| | - Samuel T Workenhe
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Prathyusha Konda
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Shashi Gujar
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada; Department of Pathology, Dalhousie University, Halifax, NS, Canada; Department of Biology, Dalhousie University, Halifax, NS, Canada; Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe 11 labellisée par la Ligue Nationale contre le Cancer, INSERM, Sorbonne Université, Université de Paris, Paris, France; Gustave Roussy Cancer Campus, Metabolomics and Cell Biology Platforms, Villejuif, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France; Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China; Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
10
|
Geoffroy K, Bourgeois-Daigneault MC. The pros and cons of interferons for oncolytic virotherapy. Cytokine Growth Factor Rev 2020; 56:49-58. [PMID: 32694051 DOI: 10.1016/j.cytogfr.2020.07.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 07/02/2020] [Indexed: 12/29/2022]
Abstract
Interferons (IFN) are potent immune stimulators that play key roles in both innate and adaptive immune responses. They are considered the first line of defense against viral pathogens and can even be used as treatments to boost the immune system. While viruses are usually seen as a threat to the host, an emerging class of cancer therapeutics exploits the natural capacity of some viruses to directly infect and kill cancer cells. The cancer-specificity of these bio-therapeutics, called oncolytic viruses (OVs), often relies on defective IFN responses that are frequently observed in cancer cells, therefore increasing their vulnerability to viruses compared to healthy cells. To ensure the safety of the therapy, many OVs have been engineered to further activate the IFN response. As a consequence of this IFN over-stimulation, the virus is cleared faster by the immune system, which limits direct oncolysis. Importantly, the therapeutic activity of OVs also relies on their capacity to trigger anti-tumor immunity and IFNs are key players in this aspect. Here, we review the complex cancer-virus-anti-tumor immunity interplay and discuss the diverse functions of IFNs for each of these processes.
Collapse
Affiliation(s)
- Karen Geoffroy
- Cancer axis and Institut du cancer de Montréal, Centre de recherche du CHUM- CRCHUM, 900 St-Denis Street, Viger Tower, Room R10.480, Montreal, Quebec, H2X0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Faculty of Medicine, Université de Montréal, 2900 Edouard-Montpetit Boulevard, Roger-Gaudry Building, Montreal, Quebec, H3T1J4, Canada
| | - Marie-Claude Bourgeois-Daigneault
- Cancer axis and Institut du cancer de Montréal, Centre de recherche du CHUM- CRCHUM, 900 St-Denis Street, Viger Tower, Room R10.480, Montreal, Quebec, H2X0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Faculty of Medicine, Université de Montréal, 2900 Edouard-Montpetit Boulevard, Roger-Gaudry Building, Montreal, Quebec, H3T1J4, Canada.
| |
Collapse
|
11
|
Yoon AR, Hong J, Li Y, Shin HC, Lee H, Kim HS, Yun CO. Mesenchymal Stem Cell-Mediated Delivery of an Oncolytic Adenovirus Enhances Antitumor Efficacy in Hepatocellular Carcinoma. Cancer Res 2019; 79:4503-4514. [PMID: 31289131 DOI: 10.1158/0008-5472.can-18-3900] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 05/01/2019] [Accepted: 07/02/2019] [Indexed: 12/27/2022]
Abstract
Oncolytic virotherapy is a promising alternative to conventional treatment, yet systemic delivery of these viruses to tumors remains a major challenge. In this regard, mesenchymal stem cells (MSC) with well-established tumor-homing property could serve as a promising systemic delivery tool. We showed that MSCs could be effectively infected by hepatocellular carcinoma (HCC)-targeted oncolytic adenovirus (HCC-oAd) through modification of the virus' fiber domain and that the virus replicated efficiently in the cell carrier. HCC-targeting oAd loaded in MSCs (HCC-oAd/MSC) effectively lysed HCC cells in vitro under both normoxic and hypoxic conditions as a result of the hypoxia responsiveness of HCC-oAd. Importantly, systemically administered HCC-oAd/MSC, which were initially infected with a low viral dose, homed to HCC tumors and resulted in a high level of virion accumulation in the tumors, ultimately leading to potent tumor growth inhibition. Furthermore, viral dose reduction and tumor localization of HCC-oAd/MSC prevented the induction of hepatotoxicity by attenuating HCC-oAd hepatic accumulation. Taken together, these results demonstrate that MSC-mediated systemic delivery of oAd is a promising strategy for achieving synergistic antitumor efficacy with improved safety profiles. SIGNIFICANCE: Mesenchymal stem cells enable delivery of an oncolytic adenovirus specifically to the tumor without posing any risk associated with systemic administration of naked virions to the host.
Collapse
Affiliation(s)
- A-Rum Yoon
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea (South).,Institute of Nano Science and Technology (INST), Hanyang University, Seoul, Republic of Korea (South)
| | - JinWoo Hong
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea (South).,GeneMedicine CO., Ltd., Seoul, Republic of Korea (South)
| | - Yan Li
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea (South)
| | - Ha Chul Shin
- Pharmicell Co., Ltd., Gangnam-gu, Seoul, Republic of Korea (South)
| | - Hyunah Lee
- Pharmicell Co., Ltd., Gangnam-gu, Seoul, Republic of Korea (South)
| | - Hyun Soo Kim
- Pharmicell Co., Ltd., Gangnam-gu, Seoul, Republic of Korea (South)
| | - Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea (South). .,Institute of Nano Science and Technology (INST), Hanyang University, Seoul, Republic of Korea (South).,GeneMedicine CO., Ltd., Seoul, Republic of Korea (South)
| |
Collapse
|
12
|
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
|
13
|
Zhang T, Suryawanshi YR, Woyczesczyk HM, Essani K. Targeting Melanoma with Cancer-Killing Viruses. Open Virol J 2017; 11:28-47. [PMID: 28567163 PMCID: PMC5420172 DOI: 10.2174/1874357901711010028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 01/05/2017] [Accepted: 01/17/2017] [Indexed: 12/20/2022] Open
Abstract
Melanoma is the deadliest skin cancer with ever-increasing incidence. Despite the development in diagnostics and therapies, metastatic melanoma is still associated with significant morbidity and mortality. Oncolytic viruses (OVs) represent a class of novel therapeutic agents for cancer by possessing two closely related properties for tumor reduction: virus-induced lysis of tumor cells and induction of host anti-tumor immune responses. A variety of viruses, either in "natural" or in genetically modified forms, have exhibited a remarkable therapeutic efficacy in regressing melanoma in experimental and/or clinical studies. This review provides a comprehensive summary of the molecular and cellular mechanisms of action of these viruses, which involve manipulating and targeting the abnormalities of melanoma, and can be categorized as enhancing viral tropism, targeting the tumor microenvironment and increasing the innate and adaptive antitumor responses. Additionally, this review describes the "biomarkers" and deregulated pathways of melanoma that are responsible for melanoma initiation, progression and metastasis. Advances in understanding these abnormalities of melanoma have resulted in effective targeted and immuno-therapies, and could potentially be applied for engineering OVs with enhanced oncolytic activity in future.
Collapse
Affiliation(s)
- Tiantian Zhang
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, U.S.A
| | - Yogesh R. Suryawanshi
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, U.S.A
| | - Helene M. Woyczesczyk
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, U.S.A
| | - Karim Essani
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, U.S.A
| |
Collapse
|
14
|
Capitalizing on Cancer Specific Replication: Oncolytic Viruses as a Versatile Platform for the Enhancement of Cancer Immunotherapy Strategies. Biomedicines 2016; 4:biomedicines4030021. [PMID: 28536388 PMCID: PMC5344262 DOI: 10.3390/biomedicines4030021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 08/16/2016] [Accepted: 08/16/2016] [Indexed: 02/07/2023] Open
Abstract
The past decade has seen considerable excitement in the use of biological therapies in treating neoplastic disease. In particular, cancer immunotherapy and oncolytic virotherapy have emerged as two frontrunners in this regard with the first FDA approvals for agents in both categories being obtained in the last 5 years. It is becoming increasingly apparent that these two approaches are not mutually exclusive and that much of the therapeutic benefit obtained from the use of oncolytic viruses (OVs) is in fact the result of their immunotherapeutic function. Indeed, OVs have been shown to recruit and activate an antitumor immune response and much of the current work in this field centers around increasing this activity through strategies such as engineering genes for immunomodulators into OV backbones. Because of their broad immunostimulatory functions, OVs can also be rationally combined with a variety of other immunotherapeutic approaches including cancer vaccination strategies, adoptive cell transfer and checkpoint blockade. Therefore, while they are important therapeutics in their own right, the true power of OVs may lie in their ability to enhance the effectiveness of a wide range of immunotherapies.
Collapse
|
15
|
Tsun A, Miao XN, Wang CM, Yu DC. Oncolytic Immunotherapy for Treatment of Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 909:241-83. [PMID: 27240460 DOI: 10.1007/978-94-017-7555-7_5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Immunotherapy entails the treatment of disease by modulation of the immune system. As detailed in the previous chapters, the different modes of achieving immune modulation are many, including the use of small/large molecules, cellular therapy, and radiation. Oncolytic viruses that can specifically attack, replicate within, and destroy tumors represent one of the most promising classes of agents for cancer immunotherapy (recently termed as oncolytic immunotherapy). The notion of oncolytic immunotherapy is considered as the way in which virus-induced tumor cell death (known as immunogenic cancer cell death (ICD)) allows the immune system to recognize tumor cells and provide long-lasting antitumor immunity. Both immune responses toward the virus and ICD together contribute toward successful antitumor efficacy. What is now becoming increasingly clear is that monotherapies, through any of the modalities detailed in this book, are neither sufficient in eradicating tumors nor in providing long-lasting antitumor immune responses and that combination therapies may deliver enhanced efficacy. After the rise of the genetic engineering era, it has been possible to engineer viruses to harbor combination-like characteristics to enhance their potency in cancer immunotherapy. This chapter provides a historical background on oncolytic virotherapy and its future application in cancer immunotherapy, especially as a combination therapy with other treatment modalities.
Collapse
Affiliation(s)
- A Tsun
- Innovent Biologics, Inc., 168 Dongping Street, Suzhou Industrial Park, 215123, China
| | - X N Miao
- Innovent Biologics, Inc., 168 Dongping Street, Suzhou Industrial Park, 215123, China
| | - C M Wang
- Innovent Biologics, Inc., 168 Dongping Street, Suzhou Industrial Park, 215123, China
| | - D C Yu
- Innovent Biologics, Inc., 168 Dongping Street, Suzhou Industrial Park, 215123, China.
| |
Collapse
|
16
|
Liu X, Zhou Y, Liu X, Peng A, Gong H, Huang L, Ji K, Petersen RB, Zheng L, Huang K. MPHOSPH1: a potential therapeutic target for hepatocellular carcinoma. Cancer Res 2014; 74:6623-34. [PMID: 25269478 DOI: 10.1158/0008-5472.can-14-1279] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
MPHOSPH1 is a critical kinesin protein that functions in cytokinesis. Here, we show that MPHOSPH1 is overexpressed in hepatocellular carcinoma (HCC) cells, where it is essential for proliferation. Attenuating MPHOSPH1 expression with a tumor-selective shRNA-expressing adenovirus (Ad-shMPP1) was sufficient to arrest HCC cell proliferation in a manner associated with an accumulation of multinucleated polyploid cells, induction of postmitotic apoptosis, and increased sensitivity to taxol cytotoxicity. Mechanistic investigations showed that attenuation of MPHOSPH1 stabilized p53, blocked STAT3 phosphorylation, and prolonged mitotic arrest. In a mouse subcutaneous xenograft model of HCC, tumoral injection of Ad-shMPP1 inhibited MPHOSPH1 expression and tumor growth in a manner correlated with induction of apoptosis. Combining Ad-shMPP1 injection with taxol administration enhanced antitumor efficacy relative to taxol alone. Furthermore, Ad-shMPP1 tail vein injection suppressed formation of orthotopic liver nodules and prevented hepatic dysfunction. Taken together, our results identify MPHOSPH1 as an oncogenic driver and candidate therapeutic target in HCC.
Collapse
Affiliation(s)
- Xinran Liu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, China. Centre for Biomedicine Research, Wuhan Institute of Biotechnology, Wuhan, China
| | - Yafan Zhou
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyuan Liu
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | | | - Hao Gong
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, China
| | - Lizi Huang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, China
| | - Kaige Ji
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, China
| | - Robert B Petersen
- Departments of Pathology, Neuroscience and Neurology, Case Western Reserve University, Cleveland, Ohio
| | - Ling Zheng
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Kun Huang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, China. Centre for Biomedicine Research, Wuhan Institute of Biotechnology, Wuhan, China.
| |
Collapse
|
17
|
Abstract
Recent clinical data have emphatically shown the capacity of our immune systems to eradicate even advanced cancers. Although oncolytic viruses (OVs) were originally designed to function as tumour-lysing therapeutics, they have now been clinically shown to initiate systemic antitumour immune responses. Cell signalling pathways that are activated and promote the growth of tumour cells also favour the growth and replication of viruses within the cancer. The ability to engineer OVs that express immune-stimulating 'cargo', the induction of immunogenic tumour cell death by OVs and the selective targeting of OVs to tumour beds suggests that they are the ideal reagents to enhance antitumour immune responses. Coupling of OV therapy with tumour antigen vaccination, immune checkpoint inhibitors and adoptive cell therapy seems to be ready to converge towards a new generation of multimodal therapeutics to improve outcomes for cancer patients.
Collapse
Affiliation(s)
- Brian D Lichty
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S4K1, Canada
| | | | - David F Stojdl
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario (CHEO) Research Institute, Ottawa, Ontario K1H 8L1, Canada
| | - John C Bell
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada; and the Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
18
|
Abstract
Oncolytic viruses are ideal platforms for tumor vaccination because they can mediate the direct in situ killing of tumor cells that release a broad array of tumor antigens and alarmins or danger signals thereby cross-priming antitumor cytotoxic T lymphocytes (CTLs), which mediate the indirect killing of uninfected cells. The balance between the direct and indirect killing phases of oncolytic virotherapy is the key to its success and can be manipulated by incorporating various immunomodulatory genes into the oncolytic virus genome. Recently, the interim analysis of a large multicenter Phase III clinical trial for Talimogene laherparepvec, a granulocyte-macrophage colony stimulating factor-armed oncolytic herpes simplex virus, revealed significant improvement in objective response and durable response rates over control arm and a trend toward improved overall survival. Meanwhile, newer oncolytics are being developed expressing additional immunomodulatory transgenes to further enhance cross-priming and the generation of antitumor CTLs and to block the immunosuppressive actions of the tumor microenvironment. Since oncolytic vaccines can be engineered to kill tumor cells directly, modulate the kinetics of the antitumor immune response and reverse the immunosuppressive actions of the tumor, they are predicted to emerge as the preferred immunotherapeutic anticancer weapons of the future.
Collapse
Affiliation(s)
- Noura B Elsedawy
- Department of Molecular Medicine, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA
| | | |
Collapse
|
19
|
Altomonte J, Ebert O. Sorting Out Pandora's Box: Discerning the Dynamic Roles of Liver Microenvironment in Oncolytic Virus Therapy for Hepatocellular Carcinoma. Front Oncol 2014; 4:85. [PMID: 24795862 PMCID: PMC4001031 DOI: 10.3389/fonc.2014.00085] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 04/07/2014] [Indexed: 12/12/2022] Open
Abstract
Oncolytic viral therapies have recently found their way into clinical application for hepatocellular carcinoma (HCC), a disease with limited treatment options and poor prognosis. Adding to the many intrinsic challenges of in vivo oncolytic viral therapy, is the complex microenvironment of the liver, which imposes unique limitations to the successful delivery and propagation of the virus. The normal liver milieu is characterized by an intricate network of hepatocytes and non-parenchymal cells including Kupffer cells, stellate cells, and sinusoidal endothelial cells, which can secrete anti-viral cytokines, provide a platform for non-specific uptake, and form a barrier to efficient viral spread. In addition, natural killer cells are greatly enriched in the liver, contributing to the innate defense against viruses. The situation is further complicated when HCC arises in the setting of underlying hepatitis virus infection and/or hepatic cirrhosis, which occurs in more than 90% of clinical cases. These conditions pose further inhibitory effects on oncolytic virus (OV) therapy due to the presence of chronic inflammation, constitutive cytokine expression, altered hepatic blood flow, and extracellular matrix deposition. In addition, OVs can modulate the hepatic microenvironment, resulting in a complex interplay between virus and host. The immune system undoubtedly plays a substantial role in the outcome of OV therapy, both as an inhibitor of viral replication, and as a potent mechanism of virus-mediated tumor cell killing. This review will discuss the particular challenges of oncolytic viral therapy for HCC, as well as some potential strategies for modulating the immune system and synergizing with the hepatic microenvironment to improve therapeutic outcome.
Collapse
Affiliation(s)
- Jennifer Altomonte
- II. Medizinische Klinik und Poliklinik, Klinikum Rechts der Isar, Technische Universität München , München , Germany
| | - Oliver Ebert
- II. Medizinische Klinik und Poliklinik, Klinikum Rechts der Isar, Technische Universität München , München , Germany
| |
Collapse
|
20
|
Shan YF, Fang YF, Wang XQ, Jin R, Zhang QY, Andersson R. Experimental studies on treatment of pancreatic cancer with double-regulated duplicative adenovirus AdTPHre-hEndo carrying human endostatin gene. Pancreatology 2013; 13:393-400. [PMID: 23890138 DOI: 10.1016/j.pan.2013.05.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 04/27/2013] [Accepted: 05/31/2013] [Indexed: 12/11/2022]
Abstract
BACKGROUND Gene-virus targeted therapy is a promising new method of treating pancreatic cancer. To increase the efficacy and decrease the side-effect, we constructed a conditionally replicative adenovirus (CRAd) expressing human endostatin, with a human Telomoerase Reverse Transcriptase (hTERT) promoter for the regulation of the early stage of adenovirus expression of gene E1a and a Hypoxia Response Element (HRE) promoter to regulate the gene E1b. METHODS A gene recombination technique was adopted to construct and generate the adenovirus AdTPHre-hEndo. Pancreatic cancer cells were studied both in vitro and in vivo. Western blotting was adopted to observe the expressions of protein E1A and E1B; duplication assay was applied to observe the selective duplication capability of recombinant cells. MTT assay was applied to measure the lethal effects of virus on pancreatic cancer cells, and ELISA was adopted to detect the human endostatin gene expression. A pancreatic cancer transplantation tumor model of nude mice was constructed to observe the antitumor effects of the virus. RESULTS Double-regulated duplicative adenovirus AdTPHre-hEndo genes were successfully constructed. Duplication and lethal assays proved that AdTPHre-hEndo could replicate specifically in pancreatic cancer cells and kill them. The endostatin expression in a cultured supernatant from tumor cells was significantly higher than that obtained from non-duplicative adenovirus vectors carrying that gene. The animal experiment demonstrated that AdTPHre-hEndo has a high capability to limit pancreatic cancer growth. CONCLUSIONS AdTPHre-hEndo has a special ability to duplicate and kill pancreatic cancer cells in in vitro and in vivo experiments, thus providing a new gene-virus-based treatment system for pancreatic cancer.
Collapse
Affiliation(s)
- Yun-feng Shan
- Department of Surgery, First Affiliated Hospital, Wenzhou Medical College, Wenzhou 325000, Zhejiang Province, China
| | | | | | | | | | | |
Collapse
|
21
|
Das SK, Sarkar S, Dash R, Dent P, Wang XY, Sarkar D, Fisher PB. Chapter One---Cancer terminator viruses and approaches for enhancing therapeutic outcomes. Adv Cancer Res 2013; 115:1-38. [PMID: 23021240 DOI: 10.1016/b978-0-12-398342-8.00001-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
No single or combinatorial therapeutic approach has proven effective in decreasing morbidity or engendering a cure of metastatic cancer. In principle, conditionally replication-competent adenoviruses that induce tumor oncolysis through cancer-specific replication hold promise for cancer therapy. However, a single-agent approach may not be adequate to completely eradicate cancer in a patient because most cancers arise from abnormalities in multiple genetic and signal transduction pathways and targeting disseminated metastases is difficult to achieve. Based on these considerations, a novel class of cancer destroying adenoviruses have been produced, cancer terminator viruses (CTVs), in which cancer-specific replication is controlled by the progression-elevated gene-3 promoter and replicating viruses produce a second transgene encoding an apoptosis-inducing and immunomodulatory cytokine, either melanoma differentiation-associated gene-7/interleukin-24 (mda-7/IL-24) or interferon-γ. This review focuses on these viruses and ways to improve their delivery systemically and enhance their therapeutic efficacy.
Collapse
Affiliation(s)
- Swadesh K Das
- Department of Human and Molecular Genetics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Choi IK, Yun CO. Recent developments in oncolytic adenovirus-based immunotherapeutic agents for use against metastatic cancers. Cancer Gene Ther 2013; 20:70-6. [PMID: 23306610 DOI: 10.1038/cgt.2012.95] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Recurrent or metastatic cancer in most cases remains an incurable disease, and thus alternative treatment strategies, such as oncolytic virotherapy, are of great interest for clinical application. Oncolytic adenoviruses (Ads) have many advantages as virotherapeutic agents and have been safely employed in the clinics. However, the efficacy of oncolytic Ads is insufficient to eradicate tumors and current clinical applications are restricted to local administration against primary tumors because of immunological obstacles and poor tumor-cell targeting. Thus, alternative viable approaches are needed to establish therapies based on oncolytic Ad that will eliminate both primary and metastatic cancers. To this end, rational design of oncolytic Ads that express immunostimulatory genes has been employed. Even when restricted to local viral delivery, these oncolytic Ad-based immunotherapeutics have been shown to exert systemic antitumor immunity and result in eradication of both primary and metastatic cancers. Moreover, oncolytic Ad-based immunotherapeutics in combination with either dendritic cell-based vaccine or radiotherapy further strengthen the systemic tumor-specific immunity, resulting in complete suppression of both local and distant tumor metastatic growth. This review will focus on the most recent updates in strategies to develop potent oncolytic Ad-based immunotherapeutics for use in cancer gene therapy.
Collapse
Affiliation(s)
- I-K Choi
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Korea
| | | |
Collapse
|
23
|
Dong X, Qu W, Ma S, Zhu Z, Zheng C, He A, Karlsson A, Xu K, Zheng X. Potent antitumoral effects of targeted promoter-driven oncolytic adenovirus armed with Dm-dNK for breast cancer in vitro and in vivo. Cancer Lett 2012; 328:95-103. [PMID: 23000515 DOI: 10.1016/j.canlet.2012.09.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 08/20/2012] [Accepted: 09/07/2012] [Indexed: 10/27/2022]
Abstract
Currently, no curative treatments are available for late-stage metastatic or recurrent breast cancer, because the cancer tolerates both chemotherapy and endocrine therapy. In this study, we investigated the feasibility of a dual-regulated oncolytic adenoviral vector with a novel suicide gene to treat breast cancer. Following targeted gene virotherapy of conditionally replicating adenoviruses (CRAds), the novel suicide gene of multisubstrate deoxyribonucleoside kinase of Drosophila melanogaster (Dm-DNK) was inserted into the double-regulated oncolytic adenovirus SG500 to ensure more safety and enhanced antitumor activity against breast cancer both in vitro and in vivo. Selective replication, cell-killing efficacy, and cytotoxicity, combined with chemotherapeutics were investigated in several breast cell lines (MDA-MB-231 and MCF-7), normal cells (WI-38 and MRC-5), and human (MDA-MB-231) tumor models in vivo. The double-regulated SG500-dNK had high cell-killing activity in breast cancer. Replication was similar to wild-type in breast cells and was attenuated in normal cells. SG500-dNK combined with the chemotherapeutics (E)-5-(2-bromovinyl)-2'-deoxyuridine (Bvdu) and 2',2'-difluoro-deoxycytidine (dFdC) resulted in synergistically enhanced cell killing and greatly improved antitumor efficacy in vitro or in breast xenografts in vivo. These data suggest that the novel oncolytic variant SG500-dNK is a promising candidate for targeting breast tumors specifically when combined with chemotherapeutics.
Collapse
Affiliation(s)
- Xiaoshen Dong
- Department of Surgical Oncology, First Affiliated Hospital, China Medical University, Shenyang, China
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Svyatchenko VA, Tarasova MV, Netesov SV, Chumakov PM. Oncolytic adenoviruses in anticancer therapy: Current status and prospects. Mol Biol 2012. [DOI: 10.1134/s0026893312040103] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
25
|
CEA promoter-regulated oncolytic adenovirus-mediated Hsp70 expression in immune gene therapy for pancreatic cancer. Cancer Lett 2012; 319:154-163. [PMID: 22261331 DOI: 10.1016/j.canlet.2012.01.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Revised: 12/19/2011] [Accepted: 01/08/2012] [Indexed: 12/12/2022]
Abstract
Gene therapy is an important means for the comprehensive treatment of pancreatic cancer. Challenges associated with gene therapy include control of vector security and effective genetic screening. In this paper, a CEA promoter-regulated oncolytic adenovirus vector was constructed. The reporter gene assay demonstrated that the viral vector was confirmed to have tumor-specific replication features. In vitro cytology studies showed that the CEA promoter regulated the proliferation of the adenovirus vector carrying the Hsp70 gene (AdCEAp-Hsp70), which significantly increased the expression levels of Hsp70 in the CEA-positive pancreatic cancer cells, resulting in an overall reduction in the survival of cancer cells. In the human pancreatic cancer Panc-1 xenograft model in immune deficient nude mice, the CEA promoter-regulated adenovirus AdCEAp-Hsp70 significantly inhibited tumor growth. In the rat pancreatic cancer DSL-6A/C1 xenograft model in rats, the viral proliferation and high expression levels of Hsp70 promoted the interstitial infiltration of CD4+, CD8+ and gamma/delta T cells into tumors, induced host secretion of the cytokines TGF-β, INF-γ, and IL-6 and had a dual anti-tumor effects that completely inhibited the growth of pancreatic cancer. The results demonstrated that the oncolytic adenovirus under the control of CEA promoter provides additional assurances regarding the safety and efficiency of cancer gene therapy. This gene therapy model improves anti-cancer efficiency and has broad applications and developmental prospects.
Collapse
|
26
|
Liu C, Sun B, An N, Tan W, Cao L, Luo X, Yu Y, Feng F, Li B, Wu M, Su C, Jiang X. Inhibitory effect of Survivin promoter-regulated oncolytic adenovirus carrying P53 gene against gallbladder cancer. Mol Oncol 2011; 5:545-54. [PMID: 22032823 DOI: 10.1016/j.molonc.2011.10.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2011] [Revised: 10/11/2011] [Accepted: 10/11/2011] [Indexed: 12/21/2022] Open
Abstract
Gene therapy has become an important strategy for treatment of malignancies, but problems remains concerning the low gene transferring efficiency, poor transgene expression and limited targeting specific tumors, which have greatly hampered the clinical application of tumor gene therapy. Gallbladder cancer is characterized by rapid progress, poor prognosis, and aberrantly high expression of Survivin. In the present study, we used a human tumor-specific Survivin promoter-regulated oncolytic adenovirus vector carrying P53 gene, whose anti-cancer effect has been widely confirmed, to construct a wide spectrum, specific, safe, effective gene-viral therapy system, AdSurp-P53. Examining expression of enhanced green fluorecent protein (EGFP), E1A and the target gene P53 in the oncolytic adenovirus system validated that Survivin promoter-regulated oncolytic adenovirus had high proliferation activity and high P53 expression in Survivin-positive gallbladder cancer cells. Our in vitro cytotoxicity experiment demonstrated that AdSurp-P53 possessed a stronger cytotoxic effect against gallbladder cancer cells and hepatic cancer cells. The survival rate of EH-GB1 cells was lower than 40% after infection of AdSurp-P53 at multiplicity of infection (MOI) = 1 pfu/cell, while the rate was higher than 90% after infection of Ad-P53 at the same MOI, demonstrating that AdSurp-P53 has a potent cytotoxicity against EH-GB1 cells. The tumor growth was greatly inhibited in nude mice bearing EH-GB1 xenografts when the total dose of AdSurp-P53 was 1 × 10(9) pfu, and terminal dUTP nick end-labeling (TUNEL) revealed that the apoptotic rate of cancer cells was (33.4 ± 8.4)%. This oncolytic adenovirus system overcomes the long-standing shortcomings of gene therapy: poor transgene expression and targeting of only specific tumors, with its therapeutic effect better than the traditional Ad-P53 therapy regimen already on market; our system might be used for patients with advanced gallbladder cancer and other cancers, who are not sensitive to chemotherapy, radiotherapy, or who lost their chance for surgical treatment.
Collapse
Affiliation(s)
- Chen Liu
- Department of Biliary Surgery, Eastern Hepatobiliary Surgical Hospital, Second Military Medical University, Shanghai 200438, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Viral delivery for gene therapy against cell movement in cancer. Adv Drug Deliv Rev 2011; 63:671-7. [PMID: 21616108 DOI: 10.1016/j.addr.2011.05.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 04/15/2011] [Accepted: 05/07/2011] [Indexed: 12/17/2022]
Abstract
Viral delivery for cancer gene therapy is a promising approach, where traditional radiotherapy or chemotherapy to limit proliferation and movement of cancer cells has met resistance. Based on the new understanding of the biology of the viral vectors, therapeutic viral vectors for cancer gene therapy have been improved for greater safety and efficacy as well as transitioned from being non-replicating to replication-competent. Traditional oncolytic vectors have focused on eliminating tumor growth, while novel vectors simultaneously target epithelial-to-mesenchymal transition (EMT) in cancer cells, which could further prevent and reverse the aggressive tumor progression. In this review, we highlight the illustrative examples of cancer gene therapy in clinical trials as well as preclinical data and include proposals on methods to further enhance the safety and efficacy of oncolytic viral vectors in cancer gene therapy.
Collapse
|
28
|
Jiang G, Xin Y, Zheng JN, Liu YQ. Combining conditionally replicating adenovirus-mediated gene therapy with chemotherapy: a novel antitumor approach. Int J Cancer 2011; 129:263-74. [PMID: 21509783 DOI: 10.1002/ijc.25948] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Accepted: 01/05/2011] [Indexed: 12/16/2022]
Abstract
Despite significant improvements in diagnosis and innovations in the therapy of specific cancers, effective treatment of neoplastic diseases still presents major challenges. Recent studies have shown that conditionally replicating adenoviruses (CRAds) not only have the ability to destroy cancer cells but may also be potential vectors for the expression of therapeutic genes. Several studies in animal models have demonstrated that the combination of CRAds-mediated gene therapy and chemotherapy has greater therapeutic benefit than either treatment modality alone. In this review, an overview of specifications for a novel antitumor approach combining CRAd-gene therapy and chemotherapy is provided and recent progress in this field is discussed.
Collapse
Affiliation(s)
- Guan Jiang
- Center for Disease Control and Prevention of Xuzhou City, Xuzhou 221006, China
| | | | | | | |
Collapse
|
29
|
He X, Liu J, Yang C, Su C, Zhou C, Zhang Q, Li L, Wu H, Liu X, Wu M, Qian Q. 5/35 fiber-modified conditionally replicative adenovirus armed with p53 shows increased tumor-suppressing capacity to breast cancer cells. Hum Gene Ther 2011; 22:283-92. [PMID: 20846024 DOI: 10.1089/hum.2010.058] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Conditionally replicative adenoviruses (CRAds) are widely used for cancer biotherapy and show a significant growth-suppressing effect on many types of cancer. However, it was reported that breast cancer was highly resistant to the infection of traditionally used adenovirus of serotype 5 (Ad5)-based CRAds. Although partial substitution of the fiber protein of replication-deficient Ad5 with that of adenovirus of serotype 35 (Ad35) facilitated infection of breast cancer cells by adenoviral vectors, it is still unknown whether this modification can improve CRAds in their tumor-eliminating capacity. We generated a 5/35 fiber-modified CRAd with a p53 cDNA construct and investigated whether this alteration in fiber region can make CRAds suppress the growth of breast cancer more effectively. Our data reinforced the proposal that 5/35-modified fiber conferred higher adenovirus infectivity for breast cancer cells than natural Ad5 fiber. Interestingly, 5/35 fiber-modified CRAd replicated more efficiently in breast cancer cells than Ad5-based CRAd. We also found 5/35 fiber-modified CRAd mediated higher expression of p53 in breast cancer cells. In vitro, 5/35 fiber-modified CRAd eliminated breast cancer cells more efficiently. Growth of xenograft tumors in nude mice was also significantly retarded by 5/35 fiber-modified CRAd. The 5/35 fiber-modified CRAd suppressed the growth of breast cancer cells more effectively than Ad5-based CRAd, both in vitro and in vivo. Thus CRAd with 5/35 hybrid fiber may be a promising vector for breast cancer treatment.
Collapse
Affiliation(s)
- Xiaoping He
- Laboratory of Gene and Viral Therapy, Eastern Hepatobiliary Surgical Hospital, The Second Military Medical University, Shanghai 200438, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Increasing the efficacy of oncolytic adenovirus vectors. Viruses 2010; 2:1844-1866. [PMID: 21994711 PMCID: PMC3185754 DOI: 10.3390/v2091844] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Revised: 08/17/2010] [Accepted: 08/25/2010] [Indexed: 12/13/2022] Open
Abstract
Oncolytic adenovirus (Ad) vectors present a new modality to treat cancer. These vectors attack tumors via replicating in and killing cancer cells. Upon completion of the vector replication cycle, the infected tumor cell lyses and releases progeny virions that are capable of infecting neighboring tumor cells. Repeated cycles of vector replication and cell lysis can destroy the tumor. Numerous Ad vectors have been generated and tested, some of them reaching human clinical trials. In 2005, the first oncolytic Ad was approved for the treatment of head-and-neck cancer by the Chinese FDA. Oncolytic Ads have been proven to be safe, with no serious adverse effects reported even when high doses of the vector were injected intravenously. The vectors demonstrated modest anti-tumor effect when applied as a single agent; their efficacy improved when they were combined with another modality. The efficacy of oncolytic Ads can be improved using various approaches, including vector design, delivery techniques, and ancillary treatment, which will be discussed in this review.
Collapse
|
31
|
Bridle BW, Stephenson KB, Boudreau JE, Koshy S, Kazdhan N, Pullenayegum E, Brunellière J, Bramson JL, Lichty BD, Wan Y. Potentiating cancer immunotherapy using an oncolytic virus. Mol Ther 2010; 18:1430-9. [PMID: 20551919 DOI: 10.1038/mt.2010.98] [Citation(s) in RCA: 126] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Oncolytic viruses (OVs) are highly immunogenic and this limits their use in immune-competent hosts. Although immunosuppression may improve viral oncolysis, this gain is likely achieved at the cost of antitumoral immunity. We have developed a strategy wherein the immune response against the OV leads to enhanced therapeutic outcomes. We demonstrate that immunization with an adenoviral (Ad) vaccine before treatment with an oncolytic vesicular stomatitis virus (VSV) expressing the same tumor antigen (Ag) leads to significantly enhanced antitumoral immunity. Intratumoral replication of VSV was minimally attenuated in Ad-immunized hosts but extending the interval between treatments reduced the attenuating effect and further increased antitumoral immunity. More importantly, our combination approach shifted the immune response from viral Ags to tumor Ags and further reduced OV replication in normal tissues, leading to enhancements in both efficacy and safety. These studies also highlight the benefits of using a replicating, OV to boost a pre-existing antitumoral immune response as this approach generated larger responses versus tumor Ag in tumor-bearing hosts than could be achieved in tumor-free hosts. This strategy should be applicable to other vector combinations, tumor Ags, and tumor targets.
Collapse
Affiliation(s)
- Byram W Bridle
- Centre for Gene Therapeutics, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Toth K, Dhar D, Wold WSM. Oncolytic (replication-competent) adenoviruses as anticancer agents. Expert Opin Biol Ther 2010; 10:353-68. [PMID: 20132057 DOI: 10.1517/14712590903559822] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
IMPORTANCE OF THE FIELD Whilst therapies for neoplasies have advanced tremendously in the last few decades, there is still a need for new anti-cancer treatments. One option is genetically-engineered oncolytic adenovirus (Ad) 'vectors'. These kill cancer cells via the viral replication cycle, and amplify the anti-tumor effect by producing progeny virions able to infect neighboring tumor cells. AREAS COVERED IN THIS REVIEW We provide a description of basic Ad biology and summarize the literature for oncolytic Ads from 1996 to the present. WHAT THE READER WILL GAIN An overall view of oncolytic Ads, the merits and drawbacks of the various features of these vectors, and obstacles to further development and future directions for research. TAKE HOME MESSAGE Ads are attractive for gene therapy because they are relatively innocuous, easy to produce in large quantities, genetically stable, and easy to manipulate. A variety of have been constructed and tested, in pre-clinical and clinical experiments. Oncolytic Ads proved to be remarkably safe; no dose-limiting toxicity was observed in any clinical trial, and the maximum tolerated dose was not reached. At present, the major challenge for researchers is to increase the efficacy of the vectors, and to incorporate oncolytic virotherapy into existing treatment protocols.
Collapse
Affiliation(s)
- Karoly Toth
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA.
| | | | | |
Collapse
|
33
|
Kaur B, Cripe TP, Chiocca EA. "Buy one get one free": armed viruses for the treatment of cancer cells and their microenvironment. Curr Gene Ther 2010; 9:341-55. [PMID: 19860649 DOI: 10.2174/156652309789753329] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Oncolytic viral therapy is a promising biological therapy for the treatment of cancer. Recent advances in genetic engineering have facilitated the construction of custom-built oncolytic viruses that can be exquisitely targeted to tumors by exploiting each cancer's unique biology and their efficacy can be further enhanced by "arming" them with additional therapeutic genes. Such an approach allows the virus to unload its "therapeutic cargo" at the tumor site, thereby enhancing its anti-neoplastic properties. While several clever strategies have been recently described using genes that can induce cellular apoptosis/suicide and/or facilitate tumor/virus imaging, viruses armed with genes that also affect the tumor microenvironment present an exciting and promising approach to therapy. In this review we discuss recently developed oncolytic viruses armed with genes encoding for angiostatic factors, inflammatory cytokines, or proteases that modulate the extracellular matrix to regulate tumor vascularization, anti-tumor immune responses and viral spread throughout the solid tumor.
Collapse
Affiliation(s)
- Balveen Kaur
- Dardinger Laboratory for Neuro-oncology and Neurosciences, Department of Neurological Surgery, James Comprehensive Cancer Center and The Ohio State University Medical Center, 400 West 12th Avenue, Columbus, OH 43210,USA.
| | | | | |
Collapse
|
34
|
Ottolino-Perry K, Diallo JS, Lichty BD, Bell JC, McCart JA. Intelligent design: combination therapy with oncolytic viruses. Mol Ther 2009; 18:251-63. [PMID: 20029399 DOI: 10.1038/mt.2009.283] [Citation(s) in RCA: 149] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Metastatic cancer remains an incurable disease in the majority of cases and thus novel treatment strategies such as oncolytic virotherapy are rapidly advancing toward clinical use. In order to be successful, it is likely that some type of combination therapy will be necessary to have a meaningful impact on this disease. Although it may be tempting to simply combine an oncolytic virus with the existing standard radiation or chemotherapeutics, the long-term goal of such treatments must be to have a rational, potentially synergistic combination strategy that can be safely and easily used in the clinical setting. The combination of oncolytic virotherapy with existing radiotherapy and chemotherapy modalities is reviewed along with novel biologic therapies including immunotherapies, in order to help investigators make intelligent decisions during the clinical development of these products.
Collapse
Affiliation(s)
- Kathryn Ottolino-Perry
- Division of Experimental Therapeutics, Toronto General Research Institute, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
35
|
Park MY, Kim DR, Jung HW, Yoon HI, Lee JH, Lee CT. Genetic immunotherapy of lung cancer using conditionally replicating adenovirus and adenovirus-interferon-β. Cancer Gene Ther 2009; 17:356-64. [DOI: 10.1038/cgt.2009.78] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
36
|
Xie M, Niu JH, Chang Y, Qian QJ, Wu HP, Li LF, Zhang Y, Li JL, Huang XJ, Ruan GR. A novel triple-regulated oncolytic adenovirus carrying PDCD5 gene exerts potent antitumor efficacy on common human leukemic cell lines. Apoptosis 2009; 14:1086-94. [PMID: 19551515 DOI: 10.1007/s10495-009-0373-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PDCD5 (programmed cell death 5) accelerates apoptosis of certain tumor cells and the replication-defective Ad-PDCD5 may be a promising agent for enhancing chemosensitivity. In this study, a triple-regulated conditionally replicating adenoviruses (CRAd) carrying PDCD5 gene expression cassette, SG611-PDCD5, was engineered. In SG611-PDCD5, the E1a gene with a deletion of 24 nucleotides within CR2 region is controlled under the human telomerase reverse transcriptase (hTERT) promoter, the E1b gene expression is directed by the hypoxia response element (HRE), whereas the PDCD5 gene is controlled by the cytomegalovirus promoter. The tumor-selective replication of this virus and its antitumor efficacy were characterized in several leukemic cell lines in vitro and in xenograft models of human leukemic cell line in nude mice. It was found by RQ-RT-PCR assay that SG611-PDCD5 expressed PDCD5 efficiently in leukemic cells. In K562 tumor xenograft models, SG611-PDCD5 displayed a tumor killing capacity. At a dose of 1 x 10(9) plaque-forming units, SG611-PDCD5 alone could completely inhibit the tumor growth and more effective than replication-defective Ad-PDCD5. Histopathologic examination revealed that SG611-PDCD5 administration resulted in leukemic cell apoptosis. We concluded that the triple-regulated SG611-PDCD5, as a more potent and safer antitumor therapeutic, could provide a new strategy for leukemia biotherapy.
Collapse
Affiliation(s)
- Min Xie
- Peking University People's Hospital and Institute of Hematology, Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Fecker LF, Schmude M, Jost S, Hossini AM, Picó AH, Wang X, Schwarz C, Fechner H, Eberle J. Efficient and selective tumor cell lysis and induction of apoptosis in melanoma cells by a conditional replication-competent CD95L adenovirus. Exp Dermatol 2009; 19:e56-66. [DOI: 10.1111/j.1600-0625.2009.00977.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
38
|
Doronin K, Shashkova EV, May SM, Hofherr SE, Barry MA. Chemical modification with high molecular weight polyethylene glycol reduces transduction of hepatocytes and increases efficacy of intravenously delivered oncolytic adenovirus. Hum Gene Ther 2009; 20:975-88. [PMID: 19469693 PMCID: PMC2829283 DOI: 10.1089/hum.2009.028] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2009] [Accepted: 05/22/2009] [Indexed: 12/19/2022] Open
Abstract
Oncolytic adenoviruses are anticancer agents that replicate within tumors and spread to uninfected tumor cells, amplifying the anticancer effect of initial transduction. We tested whether coating the viral particle with polyethylene glycol (PEG) could reduce transduction of hepatocytes and hepatotoxicity after systemic (intravenous) administration of oncolytic adenovirus serotype 5 (Ad5). Conjugating Ad5 with high molecular weight 20-kDa PEG but not with 5-kDa PEG reduced hepatocyte transduction and hepatotoxicity after intravenous injection. PEGylation with 20-kDa PEG was as efficient at detargeting adenovirus from Kupffer cells and hepatocytes as virus predosing and warfarin. Bioluminescence imaging of virus distribution in two xenograft tumor models in nude mice demonstrated that PEGylation with 20-kDa PEG reduced liver infection 19- to 90-fold. Tumor transduction levels were similar for vectors PEGylated with 20-kDa PEG and unPEGylated vectors. Anticancer efficacy after a single intravenous injection was retained at the level of unmodified vector in large established prostate carcinoma xenografts, resulting in complete elimination of tumors in all animals and long-term tumor-free survival. Anticancer efficacy after a single intravenous injection was increased in large established hepatocellular carcinoma xenografts, resulting in significant prolongation of survival as compared with unmodified vector. The increase in efficacy was comparable to that obtained with predosing and warfarin pretreatment, significantly extending the median of survival. Shielding adenovirus with 20-kDa PEG may be a useful approach to improve the therapeutic window of oncolytic adenovirus after systemic delivery to primary and metastatic tumor sites.
Collapse
Affiliation(s)
- Konstantin Doronin
- Division of Infectious Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902
| | - Elena V. Shashkova
- Division of Infectious Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902
| | - Shannon M. May
- Division of Infectious Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902
| | - Sean E. Hofherr
- Division of Infectious Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902
| | - Michael A. Barry
- Division of Infectious Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902
- Department of Immunology, Mayo Clinic, Rochester, MN 55902
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55902
- Translational Immunovirology Program, Mayo Clinic, Rochester, MN 55902
- Cancer Center, Mayo Clinic, Rochester, MN 55902
| |
Collapse
|
39
|
Matthews KS, Alvarez RD, Curiel DT. Advancements in adenoviral based virotherapy for ovarian cancer. Adv Drug Deliv Rev 2009; 61:836-41. [PMID: 19422865 DOI: 10.1016/j.addr.2009.04.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2008] [Accepted: 04/28/2009] [Indexed: 12/17/2022]
Abstract
Ovarian cancer is a leading gynecologic malignancy with relatively grim survival statistics. There is a significant need for the development of new treatment options for this malignancy. The development of virotherapy as a treatment option for ovarian cancer has the potential to improve patient survival. Adenoviruses have multiple advantages as vectors for virotherapy including a well-understood structure and the ability to infect cells easily. We will outline the advances in virotherapy in the treatment of ovarian cancer, with particular attention directed toward adenoviral vectors.
Collapse
Affiliation(s)
- Kellie S Matthews
- The Division of Gynecologic Oncology, The University of Alabama at Birmingham, 619 19th Street South, OHB 534, Birmingham, AL 35213, USA.
| | | | | |
Collapse
|
40
|
Fang L, Huang Y, Hu X, Sun L, He X, Hu H, Pu Y, Cao X, Luo H, Pan S, Gu J, Su C. A truncated minimal-E1a gene with potency to support adenoviral replication mediates antitumor activity by down-regulating Neu expression and preserving Rb function. Chem Biol Interact 2009; 181:1-7. [PMID: 19523934 DOI: 10.1016/j.cbi.2009.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2009] [Revised: 05/21/2009] [Accepted: 06/02/2009] [Indexed: 10/20/2022]
Abstract
Oncolytic adenovirus is capable of infecting, replicating in and lysing cancer cells. In adenovirus infection and replication, the wild type E1a gene (wE1a) mediates various genetic events to facilitate viral replication and exert antitumor effect. To enhance its antitumor efficacy and optimize its safety, we manipulated the wE1a gene and designed a 720-bp truncated minimal-E1a (mE1a) by deletions and mutations of amino acid residues. The mE1a gene was incorporated in an adenovirus under the control of hTERT promoter, giving the vector AdDC315-mE1a. A variety of cancer cell lines infected with the virus expressed the mE1a protein and showed considerable down-regulation in Neu protein expression as compared to normal cell lines. mE1a also had a lower binding affinity to the Rb protein, preserving the Rb tumor suppressive function. The mE1a expression allowed efficient adenovirus replication with high and stable replication ratios in cancer cells (about 125- to 8500-fold higher at 48 h and 180- to 10,900-fold higher at 96 h post-infection). Further, the mE1a-supported oncolytic adenovirus induced higher cancer cell apoptosis, stronger cell cycle arrest and more effective antitumor efficacy in hepatocarcinoma xenografts in nude mice. In conclusion, the truncated minimal mE1a can act as a tumor inhibitor gene, and may be used to construct oncolytic adenovirus vectors for use in gene therapy of a variety of cancers.
Collapse
Affiliation(s)
- Lin Fang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Nanjing Normal University, Nanjing, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Ji X, Zhang J, Cheng L, Wei F, Li H, Liu X, Chen X, Li C, Wang Y, Huang Q. Oncolytic adenovirus delivering herpes simplex virus thymidine kinase suicide gene reduces the growth of human retinoblastoma in an in vivo mouse model. Exp Eye Res 2009; 89:193-9. [PMID: 19328781 DOI: 10.1016/j.exer.2009.03.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2008] [Revised: 03/17/2009] [Accepted: 03/17/2009] [Indexed: 01/25/2023]
Abstract
Oncolytic conditionally replicating adenoviruses (CRAd) can exclusively replicate in and lyse tumor cells and are therefore promising tools in cancer therapy. In this study, we combined the oncolytic potential of a CRAd with its ability to deliver a suicide gene (herpes simplex virus thymidine kinase suicide gene, HSVtk) in order to further enhance tumor cell killing in a human retinoblastoma (RB) mouse model. We could demonstrate that CRAd driven by the human telomerase reverse transcriptase (hTERT) promoter and armed with the HSV thymidine kinase suicide gene/ganciclovir (HSVtk/GCV) could very effectively reduce growth of human RB in an orthotopic nude mouse model. These findings suggest that hTERT promoter-driven CRAd in combination with HSVtk/GCV gene therapy could be a promising new approach for the treatment of RB. In addition, we found that hTERT promoter-driven CRAd replication occurred exclusively in human RB cells but not in primary human retinal pigment epithelial cells (hRPE), indicating that application of hTERT promoter-driven CRAd for the treatment of RB would be safe.
Collapse
Affiliation(s)
- Xunda Ji
- Experimental Center, The First People's Hospital, Shanghai Jiaotong University, Shanghai 200080, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Conditionally replicating adenoviruses (CRAds) have many advantages as agents for cancer virotherapy and have been safely used in human clinical trials. However, replicating adenoviruses have been limited in their ability to eliminate tumors by oncolysis. Thus, the efficacy of these agents must be improved. To this end, CRAds have been engineered to express therapeutic transgenes that exert antitumor effects independent of direct viral oncolysis. These transgenes can be expressed under native gene control elements, in which case placement within the genome determines the expression profile, or they can be controlled by exogenous promoters. The therapeutic transgenes used to arm replicating adenoviruses can be broadly classified into three groups. There are those that mediate killing of the infected cell, those that modulate the tumor microenvironment and those with immunomodulatory functions. Overall, the studies to date in animal models have shown that arming a CRAd with a rationally chosen therapeutic transgene can improve its antitumor efficacy over that of an unarmed CRAd. However, a number of obstacles must be overcome before the full potential of armed CRAds can be realized in the human clinical context. Hence, strategies are being developed to permit intravenous delivery to disseminated cancer cells, overcome the immune response and enable in vivo monitoring of the biodistribution and activity of armed CRAds.
Collapse
Affiliation(s)
- J J Cody
- Division of Human Gene Therapy, Department of Medicine, Gene Therapy Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | |
Collapse
|
43
|
Wang X, Su C, Cao H, Li K, Chen J, Jiang L, Zhang Q, Wu X, Jia X, Liu Y, Wang W, Liu X, Wu M, Qian Q. A novel triple-regulated oncolytic adenovirus carrying p53 gene exerts potent antitumor efficacy on common human solid cancers. Mol Cancer Ther 2008; 7:1598-603. [PMID: 18566230 DOI: 10.1158/1535-7163.mct-07-2429] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Conditionally replicating adenoviruses (CRAd) can replicate specifically in cancer cells and lyse them. The CRAds were widely used in the preclinical and clinical studies of cancer therapy. We hypothesize that more precisely regulated replication of CRAds may further improve the vector safety profile and enhance its antitumor efficacy. Here, a triple-regulated CRAd carrying p53 gene expression cassette, SG600-p53, was engineered. In SG600-p53, the E1a gene with a deletion of 24 nucleotides within CR2 region is controlled under the human telomerase reverse transcriptase (hTERT) promoter, the E1b gene expression is directed by the hypoxia response element (HRE), whereas the p53 gene is controlled by the cytomegalovirus promoter. The precise triple-regulation endows SG600-p53 with enhanced antitumor potential and improved safety profile. The tumor-selective replication of this virus and its antitumor efficacy were characterized in several tumor cell lines in vitro and in xenograft models of human non-small cell lung cancer in nude mice. With the selective replication and oncolysis, it was found by ELISA assay that SG600-p53 expressed p53 efficiently in cancer cells. In NCI-H1299 tumor xenograft models, SG600-p53 displayed a tumor-selective killing capacity. At a dose of 2 x 10(9) plaque-forming units, SG600-p53 could completely inhibit the tumor growth and more effective than replication-defective Ad-p53. Histopathologic examination revealed that SG600-p53 administration resulted in cancer cell apoptosis. We concluded that the triple-regulated SG600-p53, as a more potent and safer antitumor therapeutic, could provide a new strategy for cancer biotherapy.
Collapse
Affiliation(s)
- Xinghua Wang
- Laboratory of Viral and Gene Therapy, Eastern Hepatobiliary Surgical Hospital, Second Military Medical University, 225 Changhai Road, Shanghai 200438, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Su C, Cao H, Tan S, Huang Y, Jia X, Jiang L, Wang K, Chen Y, Long J, Liu X, Wu M, Wu X, Qian Q. Toxicology profiles of a novel p53-armed replication-competent oncolytic adenovirus in rodents, felids, and nonhuman primates. Toxicol Sci 2008; 106:242-50. [PMID: 18703561 DOI: 10.1093/toxsci/kfn168] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Conditionally replicating adenovirus (CRAd) has demonstrated to be safe in clinical studies. We generated a triple-regulated p53-armed CRAd, SG600-p53, in which the partially deleted E1a and E1b genes are regulated under the human telomerase reverse transcriptase promoter and the hypoxia response element. SG600-p53 was proven to be effective both in vitro and in vivo. In this study, the preclinical safety profiles of SG600-p53 in animal models were investigated. SG600-p53 had no adverse effects on mouse behavioral and nervous systems at 1.0 x 10(11) viral particles (VP)/kg, 2.0 x 10(11) VP/kg and 4.0 x 10(11) VP/kg doses, and on cat cardiovascular and respiratory systems at 2.0 x 10(10) VP/kg, 4.0 x 10(10) VP/kg, and 8.0 x 10(10) VP/kg doses. In acute toxicity test in mice, the maximum tolerated dose (2.5 x 10(13) VP/kg) induced cachexia, decreased activity, and eye closure in 9/20 mice which could be self-resolved within 30 min. Sensitized by five repeated ip injections at 1.0 x 10(10) VP/kg each ip and excitated by one iv injection at 1.0 x 10(11) VP/kg, guinea pigs did not show any sign of systemic anaphylaxis. In repeat-dose toxicological studies, the no-observable-adverse-effect levels of SG600-p53 in rats (1.0 x 10(11) VP/kg) and cynomolgus monkeys (5.0 x 10(11) VP/kg) were 12-fold and 60-fold of the proposed clinical dose, respectively. Intramuscular injections of SG600-p53 in cynomolgus monkeys caused inflammation at injection sites, which was alleviative at the end of observation period. The anti-virus antibody was produced in animal sera and decreased gradually 4 weeks later. No histopathological changes were found by bone marrow examination. Our data in different animal models suggest that SG600-p53 is a safe antitumor therapeutic agent.
Collapse
Affiliation(s)
- Changqing Su
- Laboratory of Viral and Gene Therapy, Eastern Hepatobiliary Surgical Hospital, Second Military Medical University, Shanghai 200438, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Vigil A, Park MS, Martinez O, Chua MA, Xiao S, Cros JF, Martínez-Sobrido L, Woo SLC, García-Sastre A. Use of reverse genetics to enhance the oncolytic properties of Newcastle disease virus. Cancer Res 2007; 67:8285-92. [PMID: 17804743 DOI: 10.1158/0008-5472.can-07-1025] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Naturally occurring strains of Newcastle disease virus (NDV) have shown oncolytic therapeutic efficacy in preclinical studies and are currently in clinical trials. Here, we have evaluated the possibility to enhance the cancer therapeutic potential of NDV by means of reverse genetics. Mice bearing s.c. implanted CT26 tumors were treated with intratumoral (i.t.) injections of a recombinant NDV modified to contain a highly fusogenic F protein. These treated mice exhibited significant reduction in tumor development compared with mice treated with the unmodified virus. Furthermore, mice in a CT26 metastatic tumor model treated with an i.v. injection of the genetically engineered NDV exhibited prolonged survival compared with wild-type control virus. In addition, we examined whether the oncolytic properties of NDV could be improved by expression of immunostimulatory molecules. In this regard, we engineered several NDVs to express granulocyte macrophage colony-stimulating factor, IFN-gamma, interleukin 2 (IL-2), or tumor necrosis factor alpha, and evaluated their therapeutic potential in an immunocompetent colon carcinoma tumor model. Mice bearing s.c. CT26 tumors treated with i.t. injections of recombinant NDV expressing IL-2 showed dramatic reductions in tumor growth, with a majority of the mice undergoing complete and long-lasting remission. Our data show the use of reverse genetics to develop enhanced recombinant NDV vectors as effective therapeutic agents for cancer treatment.
Collapse
Affiliation(s)
- Adam Vigil
- Department of Microbiology, Medicine, Division of Infectious Diseases, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Yang ZR, Wang HF, Zhao J, Peng YY, Wang J, Guinn BA, Huang LQ. Recent developments in the use of adenoviruses and immunotoxins in cancer gene therapy. Cancer Gene Ther 2007; 14:599-615. [PMID: 17479105 DOI: 10.1038/sj.cgt.7701054] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Despite setbacks in the past and apparent hurdles ahead, gene therapy is advancing toward reality. The past several years have witnessed this new field of biomedicine developing rapidly both in breadth and depth, especially for the treatment of cancer, thanks largely to the better understanding of molecular and genetic basis of oncogenesis and the development of new and improved vectors and technologies for gene delivery and targeting. This article is intended to provide a brief review of recent advances in cancer gene therapy using adenoviruses, both as vectors and as oncolytic agents, and some of the recent progress in the development of immunotoxins for use in cancer gene therapy.
Collapse
Affiliation(s)
- Z R Yang
- Center for Biotech & BioMedicine and Division of Life Sciences, Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | | | | | | | | | | | | |
Collapse
|
47
|
Vähä-Koskela MJ, Heikkilä JE, Hinkkanen AE. Oncolytic viruses in cancer therapy. Cancer Lett 2007; 254:178-216. [PMID: 17383089 PMCID: PMC7126325 DOI: 10.1016/j.canlet.2007.02.002] [Citation(s) in RCA: 220] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2006] [Revised: 02/01/2007] [Accepted: 02/05/2007] [Indexed: 12/26/2022]
Abstract
Oncolytic virotherapy is a promising form of gene therapy for cancer, employing nature’s own agents to find and destroy malignant cells. The purpose of this review is to provide an introduction to this very topical field of research and to point out some of the current observations, insights and ideas circulating in the literature. We have strived to acknowledge as many different oncolytic viruses as possible to give a broader picture of targeting cancer using viruses. Some of the newest additions to the panel of oncolytic viruses include the avian adenovirus, foamy virus, myxoma virus, yaba-like disease virus, echovirus type 1, bovine herpesvirus 4, Saimiri virus, feline panleukopenia virus, Sendai virus and the non-human coronaviruses. Although promising, virotherapy still faces many obstacles that need to be addressed, including the emergence of virus-resistant tumor cells.
Collapse
Affiliation(s)
- Markus J.V. Vähä-Koskela
- Åbo Akademi University, Department of Biochemistry and Pharmacy and Turku Immunology Centre, Turku, Finland
- Turku Graduate School of Biomedical Sciences, Turku, Finland
- Corresponding author. Address: Åbo Akademi University, Department of Biochemistry and Pharmacy and Turku Immunology Centre, Turku, Finland. Tel.: +358 2 215 4018; fax: +358 2 215 4745.
| | - Jari E. Heikkilä
- Åbo Akademi University, Department of Biochemistry and Pharmacy and Turku Immunology Centre, Turku, Finland
| | - Ari E. Hinkkanen
- Åbo Akademi University, Department of Biochemistry and Pharmacy and Turku Immunology Centre, Turku, Finland
| |
Collapse
|
48
|
Zhang Q, Chen G, Peng L, Wang X, Yang Y, Liu C, Shi W, Su C, Wu H, Liu X, Wu M, Qian Q. Increased safety with preserved antitumoral efficacy on hepatocellular carcinoma with dual-regulated oncolytic adenovirus. Clin Cancer Res 2007; 12:6523-31. [PMID: 17085667 DOI: 10.1158/1078-0432.ccr-06-1491] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE A dual-regulated adenovirus variant CNHK500, in which human telomerase reverse transcriptase promoter drove the adenovirus 5 (Ad5) E1a gene and hypoxia-response promoter controlled the E1b gene, was engineered. This virus has broad anticancer spectrum and higher specificity compared with mono-regulated adenovirus CNHK300. The objective of the current study is to show its antitumor selectivity and therapeutic potential. EXPERIMENTAL DESIGN The antitumor specificity of human telomerase reverse transcriptase and hypoxia response promoters was evaluated in a panel of tumor and normal cells. Under the control of these promoters, the tumor-selective expression of E1a and E1b genes was evaluated. Further in vitro antitumor specificity and potency of this virus were characterized by viral replication and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Subsequently, hepatocellular carcinoma xenografts were established to evaluate CNHK500 antitumor efficacy in vivo by different routes of virus administration and different dosages. RESULTS Human telomerase reverse transcriptase and hypoxia response promoters were activated in a tumor-selective manner or under hypoxia treatment in a broad panel of cells. Selective adenoviral early gene expression, efficient viral replication, and oncolysis were observed in all tested cancer cells with more attenuated replication capacity in normal cells. Significant regression of hepatocellular carcinoma xenografts and prolonged survival were observed by either i.t. or i.v. administration. CONCLUSIONS CNHK500 greatly reduced side effects in normal cells via dual control of adenoviral essential genes while still preserving potent antitumor efficacy on broad-spectrum cancer cells in vitro and in vivo. It can be used as a powerful therapeutic agent not only for liver cancers but also for other solid tumors.
Collapse
Affiliation(s)
- Qi Zhang
- Laboratory of Viral and Gene Therapy, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Hangzhou, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
N/A. N/A. Shijie Huaren Xiaohua Zazhi 2006; 14:1755-1761. [DOI: 10.11569/wcjd.v14.i18.1755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|