1
|
Zhao Y, Liu H, Zhan Q, Jin H, Wang Y, Wang H, Huang B, Huang F, Jia X, Wang Y, Wang X. Oncolytic adenovirus encoding LHPP exerts potent antitumor effect in lung cancer. Sci Rep 2024; 14:13108. [PMID: 38849383 PMCID: PMC11161505 DOI: 10.1038/s41598-024-63325-z] [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: 01/22/2024] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
LHPP has been shown to be a new tumor suppressor, and has a tendency to be under-expressed in a variety of cancers. Oncolytic virotheray is a promising therapeutics for lung cancer in recent decade years. Here we successfully constructed a new recombinant oncolytic adenovirus GD55-LHPP and investigated the effect of GD55-LHPP on the growth of lung cancer cells in vitro and in vivo. The results showed that LHPP had lower expression in either lung cancer cells or clinical lung cancer tissues compared with normal cells or tissues, and GD55-LHPP effectively mediated LHPP expression in lung cancer cells. GD55-LHPP could effectively inhibit the proliferation of lung cancer cell lines and rarely affected normal cell growth. Mechanically, the oncolytic adenovirus GD55-LHPP was able to induce stronger apoptosis of lung cancer cells compared with GD55 through the activation of caspase signal pathway. Notably, GD55-LHPP also activated autophagy-related signal pathway. Further, GD55-LHPP efficiently inhibited tumor growth in lung cancer xenograft in mice and prolonged animal survival rate compared with the control GD55 or PBS. In conclusion, the novel construct GD55-LHPP provides a valuable strategy for lung cancer-targeted therapy and develop the role of tumor suppress gene LHPP in lung cancer gene therapy.
Collapse
Affiliation(s)
- Yaru Zhao
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
- Oncology Department, Zhejiang Xiaoshan HospitaI, Hangzhou, China
| | - Huihui Liu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Qi Zhan
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Hao Jin
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Yiqiang Wang
- Surgical Department of Duchang County Second People's Hospital, Jiujiang, 332600, China
| | - Hui Wang
- Oncology Department, Zhejiang Xiaoshan HospitaI, Hangzhou, China
| | - Biao Huang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Fang Huang
- Department of Pathology, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China
| | - Xiaoyuan Jia
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
| | - Yigang Wang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
| | - Xiaoyan Wang
- Oncology Department, Zhejiang Xiaoshan HospitaI, Hangzhou, China.
| |
Collapse
|
2
|
Zhang J, Chen H, Chen C, Liu H, He Y, Zhao J, Yang P, Mao Q, Xia H. Systemic administration of mesenchymal stem cells loaded with a novel oncolytic adenovirus carrying IL-24/endostatin enhances glioma therapy. Cancer Lett 2021; 509:26-38. [PMID: 33819529 DOI: 10.1016/j.canlet.2021.03.027] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 12/20/2022]
Abstract
Oncolytic adenovirus-mediated gene therapy shows promise for cancer treatment; however, the systemic delivery of oncolytic adenovirus to tumors remains challenging. Recently, mesenchymal stem cells (MSCs) have emerged as potential vehicles for improving delivery. Yet, because the oncolytic adenovirus replicates in MSCs, balancing MSC viability with viral load is key to achieving optimal therapeutic effect. We thus developed an all-in-one Tet-on system that can regulate replication of oncolytic adenovirus. Then, we loaded the novel oncolytic adenovirus carrying interleukin (IL)-24 and/or Endostatin in human umbilical cord blood-mesenchymal stem cells (hUCB-MSCs) for glioma therapy. In vitro assays demonstrated that this novel oncolytic adenovirus could efficiently replicate and kill glioma cells while sparing normal cells. Moreover, doxycycline effectively regulated oncolytic adenovirus replication in the hUCB-MSCs. The doxycycline induction group with dual expression of IL-24 and Endostatin exhibited significantly greater antitumor effects than other groups in a xenograft model of glioma. Thus, this strategy for systemic delivery of oncolytic adenovirus with its oncolytic activity controlled by a Tet-on system is a promising method for achieving antitumor efficacy in glioma, especially for metastatic tumors.
Collapse
Affiliation(s)
- Junhe Zhang
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, 710062, PR China; Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang, 453003, PR China.
| | - Hao Chen
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, 710062, PR China; Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, PR China.
| | - Chen Chen
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, 710062, PR China.
| | - Haimeng Liu
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, 710062, PR China.
| | - Yurou He
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, 710062, PR China.
| | - Junli Zhao
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, 710062, PR China.
| | - Peiyan Yang
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, 710062, PR China.
| | - Qinwen Mao
- Department of Pathology, University of Utah, Huntsman Cancer Institute, 2000 Circle of Hope Drive, Salt Lake City, UT, 84112, USA.
| | - Haibin Xia
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, 710062, PR China.
| |
Collapse
|
3
|
Wang Z, Peet NP, Zhang P, Jiang Y, Rong L. Current Development of Glioblastoma Therapeutic Agents. Mol Cancer Ther 2021; 20:1521-1532. [PMID: 34172531 DOI: 10.1158/1535-7163.mct-21-0159] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/05/2021] [Accepted: 06/15/2021] [Indexed: 11/16/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive malignant primary brain tumor in humans. Over the past several decades, despite improvements in neurosurgical techniques, development of powerful chemotherapeutic agents, advances in radiotherapy, and comprehensive genomic profiling and molecular characterization, treatment of GBM has achieved very limited success in increasing overall survival. Thus, identifying and understanding the key molecules and barriers responsible for the malignant phenotypes and treatment resistance of GBM will yield new potential therapeutic targets. We review the most recent development of receptor tyrosine kinase targeted therapy for GBM and discuss the current status of several novel strategies with the emphasis on blood-brain barrier penetration as a major obstacle for small-molecule drugs to achieve their therapeutic goals. Likewise, a major opportunity for the treatment of GBM lies in the use of biomarkers for the discovery and development of new receptor tyrosine kinase targeted therapy.
Collapse
Affiliation(s)
- Zilai Wang
- Chicago BioSolutions, Inc., Chicago, Illinois.
| | | | - Pin Zhang
- Chicago BioSolutions, Inc., Chicago, Illinois
| | - Yuwei Jiang
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Lijun Rong
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois.
| |
Collapse
|
4
|
Laevskaya A, Borovjagin A, Timashev PS, Lesniak MS, Ulasov I. Metabolome-Driven Regulation of Adenovirus-Induced Cell Death. Int J Mol Sci 2021; 22:ijms22010464. [PMID: 33466472 PMCID: PMC7796492 DOI: 10.3390/ijms22010464] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/26/2020] [Accepted: 12/27/2020] [Indexed: 02/06/2023] Open
Abstract
A viral infection that involves virus invasion, protein synthesis, and virion assembly is typically accompanied by sharp fluctuations in the intracellular levels of metabolites. Under certain conditions, dramatic metabolic shifts can result in various types of cell death. Here, we review different types of adenovirus-induced cell death associated with changes in metabolic profiles of the infected cells. As evidenced by experimental data, in most cases changes in the metabolome precede cell death rather than represent its consequence. In our previous study, the induction of autophagic cell death was observed following adenovirus-mediated lactate production, acetyl-CoA accumulation, and ATP release, while apoptosis was demonstrated to be modulated by alterations in acetate and asparagine metabolism. On the other hand, adenovirus-induced ROS production and ATP depletion were demonstrated to play a significant role in the process of necrotic cell death. Interestingly, the accumulation of ceramide compounds was found to contribute to the induction of all the three types of cell death mentioned above. Eventually, the characterization of metabolite analysis could help in uncovering the molecular mechanism of adenovirus-mediated cell death induction and contribute to the development of efficacious oncolytic adenoviral vectors.
Collapse
Affiliation(s)
- Anastasia Laevskaya
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
| | - Anton Borovjagin
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Peter S. Timashev
- Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
- Department of Polymers and Composites, N.N.Semenov Institute of Chemical Physics, 4 Kosygin St., 119991 Moscow, Russia
- Chemistry Department, Lomonosov Moscow State University, Leninskiye Gory 1-3, 119991 Moscow, Russia
| | - Maciej S. Lesniak
- Department of Neurological Surgery, Northwestern University, Chicago, IL 60601, USA;
| | - Ilya Ulasov
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
- Correspondence:
| |
Collapse
|
5
|
Xiao B, Zhang L, Liu H, Fang H, Wang C, Huang B, Liu X, Zhou X, Wang Y. Oncolytic Adenovirus CD55-Smad4 Suppresses Cell Proliferation, Metastasis, and Tumor Stemness in Colorectal Cancer by Regulating Wnt/β-Catenin Signaling Pathway. Biomedicines 2020; 8:593. [PMID: 33322272 PMCID: PMC7763845 DOI: 10.3390/biomedicines8120593] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 12/08/2020] [Indexed: 12/14/2022] Open
Abstract
During the past few decades, colorectal cancer (CRC) incidence and mortality have significantly increased, and CRC has become the leading cause of cancer-related death worldwide. Thus, exploring novel effective therapies for CRC is imperative. In this study, we investigated the effect of oncolytic adenovirus CD55-Smad4 on CRC cell growth. Cell viability assay, animal experiments, flow cytometric analysis, cell migration, and invasion assays, and Western blotting were used to detect the proliferation, apoptosis, migration, and invasion of CRC cells. The oncolytic adenovirus CD55-Smad4 was successfully constructed and effectively suppressed CRC cell proliferation in vivo and in vitro. Notably, CD55-Smad4 activated the caspase signaling pathway, inducing the apoptosis of CRC cells. Additionally, the generated oncolytic adenovirus significantly suppressed migration and invasion of CRC cells by overexpressing Smad4 and inhibiting Wnt/β-catenin/epithelial-mesenchymal transition (EMT) signaling pathway. Moreover, CRC cells treated with CD55-Smad4 formed less and smaller spheroid colonies in serum-free culture than cells in control groups, suggesting that CD55-Smad4 suppressed the stemness of CRC cells by inhibiting the Wnt/β-catenin pathway. Together, the results of this study provide valuable information for the development of a novel strategy for cancer-targeting gene-virotherapy and provide a deeper understanding of the critical significance of Smad4 in gene therapy of CRC.
Collapse
Affiliation(s)
- Boduan Xiao
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (B.X.); (L.Z.); (H.L.); (H.F.); (C.W.); (B.H.); (X.L.); (X.Z.)
| | - Leilei Zhang
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (B.X.); (L.Z.); (H.L.); (H.F.); (C.W.); (B.H.); (X.L.); (X.Z.)
| | - Huihui Liu
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (B.X.); (L.Z.); (H.L.); (H.F.); (C.W.); (B.H.); (X.L.); (X.Z.)
| | - Huiling Fang
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (B.X.); (L.Z.); (H.L.); (H.F.); (C.W.); (B.H.); (X.L.); (X.Z.)
| | - Chunming Wang
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (B.X.); (L.Z.); (H.L.); (H.F.); (C.W.); (B.H.); (X.L.); (X.Z.)
| | - Biao Huang
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (B.X.); (L.Z.); (H.L.); (H.F.); (C.W.); (B.H.); (X.L.); (X.Z.)
| | - Xinyuan Liu
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (B.X.); (L.Z.); (H.L.); (H.F.); (C.W.); (B.H.); (X.L.); (X.Z.)
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiumei Zhou
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (B.X.); (L.Z.); (H.L.); (H.F.); (C.W.); (B.H.); (X.L.); (X.Z.)
| | - Yigang Wang
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (B.X.); (L.Z.); (H.L.); (H.F.); (C.W.); (B.H.); (X.L.); (X.Z.)
| |
Collapse
|
6
|
Zhang Q, Liu F. Advances and potential pitfalls of oncolytic viruses expressing immunomodulatory transgene therapy for malignant gliomas. Cell Death Dis 2020; 11:485. [PMID: 32587256 PMCID: PMC7316762 DOI: 10.1038/s41419-020-2696-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/09/2020] [Accepted: 06/15/2020] [Indexed: 12/12/2022]
Abstract
Glioblastoma (GBM) is an immunosuppressive, lethal brain tumor. Despite advances in molecular understanding and therapies, the clinical benefits have remained limited, and the life expectancy of patients with GBM has only been extended to ~15 months. Currently, genetically modified oncolytic viruses (OV) that express immunomodulatory transgenes constitute a research hot spot in the field of glioma treatment. An oncolytic virus is designed to selectively target, infect, and replicate in tumor cells while sparing normal tissues. Moreover, many studies have shown therapeutic advantages, and recent clinical trials have demonstrated the safety and efficacy of their usage. However, the therapeutic efficacy of oncolytic viruses alone is limited, while oncolytic viruses expressing immunomodulatory transgenes are more potent inducers of immunity and enhance immune cell-mediated antitumor immune responses in GBM. An increasing number of basic studies on oncolytic viruses encoding immunomodulatory transgene therapy for malignant gliomas have yielded beneficial outcomes. Oncolytic viruses that are armed with immunomodulatory transgenes remain promising as a therapy against malignant gliomas and will undoubtedly provide new insights into possible clinical uses or strategies. In this review, we summarize the research advances related to oncolytic viruses that express immunomodulatory transgenes, as well as potential treatment pitfalls in patients with malignant gliomas.
Collapse
Affiliation(s)
- Qing Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China.
- Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, 100070, China.
- Beijing Laboratory of Biomedical Materials, Beijing, 100070, China.
| | - Fusheng Liu
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China.
- Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, 100070, China.
- Beijing Laboratory of Biomedical Materials, Beijing, 100070, China.
| |
Collapse
|
7
|
González-Pastor R, Ashshi AM, El-Shemi AG, Dmitriev IP, Kashentseva EA, Lu ZH, Goedegebuure SP, Podhajcer OL, Curiel DT. Defining a murine ovarian cancer model for the evaluation of conditionally-replicative adenovirus (CRAd) virotherapy agents. J Ovarian Res 2019; 12:18. [PMID: 30767772 PMCID: PMC6376676 DOI: 10.1186/s13048-019-0493-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 02/05/2019] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Virotherapy represents a promising approach for ovarian cancer. In this regard, conditionally replicative adenovirus (CRAd) has been translated to the context of human clinical trials. Advanced design of CRAds has sought to exploit their capacity to induce anti-tumor immunization by configuring immunoregulatory molecule within the CRAd genome. Unfortunately, employed murine xenograft models do not allow full analysis of the immunologic activity linked to CRAd replication. RESULTS We developed CRAds based on the Ad5/3-Delta24 design encoding cytokines. Whereas the encoded cytokines did not impact adversely CRAd-induced oncolysis in vitro, no gain in anti-tumor activity was noted in immune-incompetent murine models with human ovarian cancer xenografts. On this basis, we explored the potential utility of the murine syngeneic immunocompetent ID8 ovarian cancer model. Of note, the ID8 murine ovarian cancer cell lines exhibited CRAd-mediated cytolysis. The use of this model now enables the rational design of oncolytic agents to achieve anti-tumor immunotherapy. CONCLUSIONS Limits of widely employed murine xenograft models of ovarian cancer limit their utility for design and study of armed CRAd virotherapy agents. The ID8 model exhibited CRAd-induced oncolysis. This feature predicate its potential utility for the study of CRAd-based virotherapy agents.
Collapse
Affiliation(s)
- Rebeca González-Pastor
- The Division of Cancer Biology and Biologic Therapeutics Center, Department of Radiation Oncology, School of Medicine, Washington University in Saint Louis, 660 South Euclid Avenue, Campus Box 8224, St. Louis, MO, 63110, USA
| | - Ahmad Mohammad Ashshi
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, PO Box 7607, Holy Makkah, Saudi Arabia
| | - Adel Galal El-Shemi
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, PO Box 7607, Holy Makkah, Saudi Arabia.,Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Igor P Dmitriev
- The Division of Cancer Biology and Biologic Therapeutics Center, Department of Radiation Oncology, School of Medicine, Washington University in Saint Louis, 660 South Euclid Avenue, Campus Box 8224, St. Louis, MO, 63110, USA
| | - Elena A Kashentseva
- The Division of Cancer Biology and Biologic Therapeutics Center, Department of Radiation Oncology, School of Medicine, Washington University in Saint Louis, 660 South Euclid Avenue, Campus Box 8224, St. Louis, MO, 63110, USA
| | - Zhi Hong Lu
- The Division of Cancer Biology and Biologic Therapeutics Center, Department of Radiation Oncology, School of Medicine, Washington University in Saint Louis, 660 South Euclid Avenue, Campus Box 8224, St. Louis, MO, 63110, USA
| | - S Peter Goedegebuure
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, 63110, USA.,Alvin J. Siteman Cancer Center, 660 S. Euclid Avenue, St. Louis, MO, 63110, USA
| | - Osvaldo L Podhajcer
- Laboratory of Molecular and Cellular Therapy, Instituto Leloir, IIBBA-CONICET, Buenos Aires, Argentina
| | - David T Curiel
- The Division of Cancer Biology and Biologic Therapeutics Center, Department of Radiation Oncology, School of Medicine, Washington University in Saint Louis, 660 South Euclid Avenue, Campus Box 8224, St. Louis, MO, 63110, USA.
| |
Collapse
|
8
|
Kaczorowski A, Hammer K, Liu L, Villhauer S, Nwaeburu C, Fan P, Zhao Z, Gladkich J, Groß W, Nettelbeck DM, Herr I. Delivery of improved oncolytic adenoviruses by mesenchymal stromal cells for elimination of tumorigenic pancreatic cancer cells. Oncotarget 2016; 7:9046-59. [PMID: 26824985 PMCID: PMC4891025 DOI: 10.18632/oncotarget.7031] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 01/15/2016] [Indexed: 01/14/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is one of the most aggressive malignancies and has poor therapeutic options. We evaluated improved oncolytic adenoviruses (OAds), in which the adenoviral gene E1B19K was deleted or a TRAIL transgene was inserted. Bone marrow mesenchymal stromal cells (MSCs) served as carriers for protected and tumor-specific virus transfers. The infection competence, tumor migration, and oncolysis were measured in cancer stem cell (CSC) models of primary and established tumor cells and in tumor xenografts. All OAds infected and lysed CSCs and prevented colony formation. MSCs migrated into PDA spheroids without impaired homing capacity. Xenotransplantation of non-infected PDA cells mixed with infected tumor cells strongly reduced the tumor volume and the expression of the proliferation marker Ki67 along with a necrotic morphology. Adenoviral capsid protein was detected in tumor xenograft tissue after intravenous injection of infected MSCs, but not in normal tissue, implying tumor-specific migration. Likewise, direct in vivo treatment correlated with a strongly reduced tumor volume, lower expression of Ki67 and CD24, and enhanced activity of caspase 3. These data demonstrate that the improved OAds induced efficient oncolysis with the OAd-TRAIL as most promising candidate for future clinical application.
Collapse
Affiliation(s)
- Adam Kaczorowski
- Surgical Research Section, Molecular OncoSurgery, Department of General and Transplantation Surgery, University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Katharina Hammer
- Oncolytic Adenovirus Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Li Liu
- Surgical Research Section, Molecular OncoSurgery, Department of General and Transplantation Surgery, University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sabine Villhauer
- Surgical Research Section, Molecular OncoSurgery, Department of General and Transplantation Surgery, University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Clifford Nwaeburu
- Surgical Research Section, Molecular OncoSurgery, Department of General and Transplantation Surgery, University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Pei Fan
- Surgical Research Section, Molecular OncoSurgery, Department of General and Transplantation Surgery, University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Zhefu Zhao
- Surgical Research Section, Molecular OncoSurgery, Department of General and Transplantation Surgery, University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jury Gladkich
- Surgical Research Section, Molecular OncoSurgery, Department of General and Transplantation Surgery, University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Wolfgang Groß
- Surgical Research Section, Molecular OncoSurgery, Department of General and Transplantation Surgery, University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dirk M Nettelbeck
- Oncolytic Adenovirus Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ingrid Herr
- Surgical Research Section, Molecular OncoSurgery, Department of General and Transplantation Surgery, University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
9
|
Naoum GE, Tawadros F, Farooqi AA, Qureshi MZ, Tabassum S, Buchsbaum DJ, Arafat W. Role of nanotechnology and gene delivery systems in TRAIL-based therapies. Ecancermedicalscience 2016; 10:660. [PMID: 27594905 PMCID: PMC4990059 DOI: 10.3332/ecancer.2016.660] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Indexed: 12/11/2022] Open
Abstract
Since its identification as a member of the tumour necrosis factor (TNF) family, TRAIL (TNF-related apoptosis-inducing ligand) has emerged as a new avenue in apoptosis-inducing cancer therapies. Its ability to circumvent the chemoresistance of conventional therapeutics and to interact with cancer stem cells (CSCs) self-renewal pathways, amplified its potential as a cancer apoptotic agent. Many recombinant preparations of this death ligand and monoclonal antibodies targeting its death receptors have been tested in monotherapy and combinational clinical trials. Gene therapy is a new approach for cancer treatment which implies viral or non-viral functional transgene induction of apoptosis in cancer cells or repair of the underlying genetic abnormality on a molecular level. The role of this approach in overcoming the traditional barriers of radiation and chemotherapeutics systemic toxicity, risk of recurrence, and metastasis made it a promising platform for cancer treatment. The recent first Food Drug Administration (FDA) approved oncolytic herpes virus for melanoma treatment brings forth the potency of the cancer gene therapy approach in the future. Many gene delivery systems have been studied for intratumoural TRAIL gene delivery alone or in combination with chemotherapeutic agents to produce synergistic cancer cytotoxicity. However, there still remain many obstacles to be conquered for this different gene delivery systems. Nanomedicine on the other hand offers a new frontier for clinical trials and biomedical research. The FDA approved nanodrugs motivates horizon exploration for other nanoscale designed particles’ implications in gene delivery. In this review we aim to highlight the molecular role of TRAIL in apoptosis and interaction with cancer stem cells (CSCs) self-renewal pathways. Finally, we also aim to discuss the different roles of gene delivery systems, mesenchymal cells, and nanotechnology designs in TRAIL gene delivery.
Collapse
Affiliation(s)
| | - Fady Tawadros
- East Tennessee State University, 1276 Gilbreath Dr, Johnson City, TN 37604, USA
| | | | | | - Sobia Tabassum
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad, Pakistan
| | - Donald J Buchsbaum
- University of Alabama at Birmingham, 1720 2nd Ave S, Birmingham, AL 35233, USA
| | - Waleed Arafat
- University of Alabama at Birmingham, 1720 2nd Ave S, Birmingham, AL 35233, USA; University of Alexandria, El-Gaish Rd, Egypt, Alexandria, Egypt
| |
Collapse
|
10
|
Ulasov IV, Shah N, Kaverina NV, Lee H, Lin B, Lieber A, Kadagidze ZG, Yoon JG, Schroeder B, Hothi P, Ghosh D, Baryshnikov AY, Cobbs CS. Tamoxifen improves cytopathic effect of oncolytic adenovirus in primary glioblastoma cells mediated through autophagy. Oncotarget 2016; 6:3977-87. [PMID: 25738357 PMCID: PMC4414167 DOI: 10.18632/oncotarget.2897] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 12/11/2014] [Indexed: 11/25/2022] Open
Abstract
Oncolytic gene therapy using viral vectors may provide an attractive therapeutic option for malignant gliomas. These viral vectors are designed in a way to selectively target tumor cells and spare healthy cells. To determine the translational impact, it is imperative to assess the factors that interfere with the anti-glioma effects of the oncolytic adenoviral vectors. In the current study, we evaluated the efficacy of survivin-driven oncolytic adenoviruses pseudotyping with adenoviral fiber knob belonging to the adenoviral serotype 3, 11 and 35 in their ability to kill glioblastoma (GBM) cells selectively without affecting normal cells. Our results indicate that all recombinant vectors used in the study can effectively target GBM in vitro with high specificity, especially the 3 knob-modified vector. Using intracranial U87 and U251 GBM xenograft models we have also demonstrated that treatment with Conditionally Replicative Adenovirus (CRAd-S-5/3) vectors can effectively regress tumor. However, in several patient-derived GBM cell lines, cells exhibited resistance to the CRAd infection as evident from the diminishing effects of autophagy. To improve therapeutic response, tumor cells were pretreated with tamoxifen. Our preliminary data suggest that tamoxifen sensitizes glioblastoma cells towards oncolytic treatment with CRAd-S-5/3, which may prove useful for GBM in future experimental therapy.
Collapse
Affiliation(s)
- Ilya V Ulasov
- Swedish Neuroscience Institute, Seattle, WA, 98122, USA.,Institute of Experimental Diagnostic and Biotherapy, NN. Blokhin Cancer Research Center, RAMN, Moscow, Russia, 115478
| | - Nameeta Shah
- Swedish Neuroscience Institute, Seattle, WA, 98122, USA
| | - Natalya V Kaverina
- NN. Blokhin Cancer Research Center, RAMN, Moscow, Russia, 115478.,Current address: Division of Nephrology, University of Washington, Seattle, 98109, USA
| | - Hwahyang Lee
- Swedish Neuroscience Institute, Seattle, WA, 98122, USA
| | - Biaoyang Lin
- Swedish Neuroscience Institute, Seattle, WA, 98122, USA
| | - Andre Lieber
- University of Washington, Seattle, WA, 98122, USA
| | | | - Jae-Guen Yoon
- Swedish Neuroscience Institute, Seattle, WA, 98122, USA
| | | | | | | | | | | |
Collapse
|
11
|
Li S, Qi Z, Li H, Hu J, Wang D, Wang X, Feng Z. Conditionally replicating oncolytic adenoviral vector expressing arresten and tumor necrosis factor-related apoptosis-inducing ligand experimentally suppresses lung carcinoma progression. Mol Med Rep 2015; 12:2068-74. [PMID: 25891208 DOI: 10.3892/mmr.2015.3624] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 01/21/2015] [Indexed: 11/06/2022] Open
Abstract
Current methods of treatment for lung carcinoma are ineffective for the majority of patients. Conditionally replicating adenoviruses (CRAds) represent a potential novel treatment for a number of neoplastic diseases, including lung carcinoma. The present study aimed to investigate the synergistic mechanisms underlying the anti-angiogenesis gene, arresten, and the apoptosis-inducing gene, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), in order to evaluate their therapeutic potential in lung cancer. The two genes were expressed by CRAd, which was confirmed using reverse transcription-polymerase chain reaction and western blotting. In vitro analyses demonstrated that CRAd adenoviruses are capable of selectively inhibiting A549 lung cancer cell growth and replication but not in that of healthy cells. In vivo analyses demonstrated that the infection of A549 cell lines using CRAd armed with the two genes (CRAd-arresten-TRAIL) enhanced the tumor inhibition, compared with cells infected with CRAd-arresten, CRAd-TRAIL or CRAd, and with the control group. CRAd-arresten-TRAIL may therefore be useful in the treatment of lung cancer.
Collapse
Affiliation(s)
- Shudong Li
- Department of Thoracic Surgery, Shaanxi Provincial People's Hospital, Third Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710068, P.R. China
| | - Zongli Qi
- Laboratory Center, Shaanxi Provincial People's Hospital, Third Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710068, P.R. China
| | - Huijin Li
- Laboratory Center, Shaanxi Provincial People's Hospital, Third Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710068, P.R. China
| | - Jun Hu
- Laboratory Center, Shaanxi Provincial People's Hospital, Third Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710068, P.R. China
| | - Dongyang Wang
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, P.R. China
| | - Xin Wang
- Laboratory Center, Shaanxi Provincial People's Hospital, Third Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710068, P.R. China
| | - Zhenzhen Feng
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, P.R. China
| |
Collapse
|
12
|
Alexiou GA, Tsamis KI, Kyritsis AP. Targeting Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL): A Promising Therapeutic Strategy in Gliomas. Semin Pediatr Neurol 2015; 22:35-9. [PMID: 25976259 DOI: 10.1016/j.spen.2014.12.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has been increasingly studied for the treatment of gliomas. TRAIL has the ability to specifically target cancer cells, without any harmful effects on normal cells, and induces apoptosis by interacting with specific receptors. Nevertheless, resistance mechanisms to TRAIL may occur at different points in the signaling pathways of TRAIL-induced apoptosis. Various approaches have been developed to overcome TRAIL resistance. Here, we have reviewed the known molecular pathways by which TRAIL exerts anticancer activity, possible resistance mechanisms, ways to sensitize resistant cancer cells, and finally the current clinical successes or limitations of TRAIL-based therapies.
Collapse
Affiliation(s)
- George A Alexiou
- Neurosurgical Institute, University of Ioannina School of Medicine, Ioannina, Greece.
| | - Konstantinos I Tsamis
- Neurosurgical Institute, University of Ioannina School of Medicine, Ioannina, Greece
| | - Athanasios P Kyritsis
- Neurosurgical Institute, University of Ioannina School of Medicine, Ioannina, Greece; Department of Neurology, University Hospital of Ioannina, Ioannina, Greece
| |
Collapse
|
13
|
Abstract
Glioblastoma Multiforme (GBM) is a rapidly progressing brain tumor. Despite the relatively low percentage of cancer patients with glioma diagnoses, recent statistics indicate that the number of glioma patients may have increased over the past decade. Current therapeutic options for glioma patients include tumor resection, chemotherapy, and concomitant radiation therapy with an average survival of approximately 16 months. The rapid progression of gliomas has spurred the development of novel treatment options, such as cancer gene therapy and oncolytic virotherapy. Preclinical testing of oncolytic adenoviruses using glioma models revealed both positive and negative sides of the virotherapy approach. Here we present a detailed overview of the glioma virotherapy field and discuss auxiliary therapeutic strategies with the potential for augmenting clinical efficacy of GBM virotherapy treatment.
Collapse
Affiliation(s)
- I.V. Ulasov
- Swedish Medical Center, Center for Advanced Brain Tumor Treatment, 550 17th Avenue, James Tower, Suite 570, Seattle, WA 98122, USA
- Institute of Experimental Diagnostic and Biotherapy, N.N. Blokhin Cancer Research Center (RONC), Moscow 115478, Russia
- Corresponding author. Ben & Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, 550 17th Avenue, James Tower, Suite 570, Seattle, WA 98122, USA. Tel.: +1 206 991 2053; fax: +1 206 834 2608.
| | - A.V. Borovjagin
- Institute of Oral Health Research, University of Alabama at Birmingham School of Dentistry, 1919 7th Ave South, Birmingham, AL, 35294, USA
| | - B.A. Schroeder
- Michigan State University College of Medicine, Grand Rapids, MI, 49503, USA
| | - A.Y. Baryshnikov
- Institute of Experimental Diagnostic and Biotherapy, N.N. Blokhin Cancer Research Center (RONC), Moscow 115478, Russia
| |
Collapse
|
14
|
Li CY, Zhang HY, Gao YL. microRNA response element‑regulated TIKI2 expression suppresses the tumorigencity of malignant gliomas. Mol Med Rep 2014; 10:2079-86. [PMID: 25051157 DOI: 10.3892/mmr.2014.2412] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 05/28/2014] [Indexed: 11/06/2022] Open
Abstract
Glioma is the most common brain malignancy and has a very poor prognosis. The current treatment options have a minimal benefit on prolonging patient survival time. Accumulating data have shown that the WNT signaling pathway has a critical function in the progression and invasion of glioma. Thus, targeting WNT signaling appears to be an effective anti‑glioma strategy. TIKI2 was recently found to suppress the activation of the WNT signaling pathway by post‑translationally modifying secreted WNT proteins. The implication of TIKI2 aberrance in cancers and its potential therapeutic effect, however, has not been studied. In the present study, a glioma‑specific adenoviral vector was constructed, which was regulated by response elements of miR‑124, to express TIKI2 in glioma cells (Ad‑TIKI2‑124). Ad‑TIKI2‑124 was found to potently suppress the activation of WNT signaling in glioma cells. This inhibitory effect on the WNT signaling pathway lead to the reduction in proliferation, colony formation ability and invasion of glioma cell lines. In addition, animal experiments confirmed that the expression of the Ad‑TIKI2‑124 construct could compromise the tumorigenicity of glioma cells in vivo. Furthermore, this glioma‑selective TIKI2 expression protected normal cells from toxicity. In conclusion, the present study demonstrated that adenovirus‑mediated TIKI2 therapy may be used for glioma treatment and therefore warrants further clinical studies.
Collapse
Affiliation(s)
- Chuan-You Li
- Department of Brain Surgery, Xinxiang Center Hospital, Xinxiang, Henan 453000, P.R. China
| | - Hong-Yun Zhang
- Department of Brain Surgery, Xinxiang Center Hospital, Xinxiang, Henan 453000, P.R. China
| | - Yi-Lu Gao
- Department of Neurosurgery, The Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
15
|
Tsamis KI, Alexiou GA, Vartholomatos E, Kyritsis AP. Combination treatment for glioblastoma cells with tumor necrosis factor-related apoptosis-inducing ligand and oncolytic adenovirus delta-24. Cancer Invest 2013; 31:630-8. [PMID: 24164301 DOI: 10.3109/07357907.2013.849724] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) exhibits cancer-selective killing activity representing a promising anticancer therapeutic strategy. Adenovirus Delta-24 is another interested anticancer agent selectively killing cells with a defective p16/Rb/E2F pathway. However, many types of cancer, including gliomas, could develop resistance to Delta-24 or TRAIL-induced apoptosis. In this study, we investigated whether TRAIL, in combination with adenovirus Delta-24, could result in an enhanced antiglioma effect in vitro in a panel of glioblastoma cell lines (U87MG, U251MG, D54, and T98G). The treatment of glioblastoma cell lines with TRAIL and Delta-24 adenovirus in combination showed markedly enhanced effect, compared to each agent alone.
Collapse
|
16
|
Abstract
Oncolytic virotherapy is a new strategy to reduce tumor burden through selective virus replication in rapidly proliferating cells. Oncolytic viruses are members of at least ten virus families, each with its advantages and disadvantages. Here, I briefly review the recent advances and key challenges, as exemplified by the best-studied platforms. Recent advances include preclinical proof of feasibility, clinical evidence of tolerability and effectiveness, and the development of new strategies to improve efficacy. These include engineered tumor selectivity and expression of antitumorigenic genes that could function independently of virus replication, identification of combinatorial therapies that accelerate intratumoral virus propagation, and modification of immune responses and vascular delivery for treatment of metastatic disease. Key challenges are to select "winners" from the distinct oncolytic platforms that can stimulate anti-cancer immunity without affecting virus replication and can lyse cancer stem cells, which are most likely responsible for tumor maintenance, aggressiveness, and recurrence. Preventing the emergence of resistant tumor cells during virotherapy through the activation of multiple death pathways, the development of a better understanding of the mechanisms of cancer stem-cell lysis, and the development of more meaningful preclinical animal models are additional challenges for the next-generation of engineered viruses.
Collapse
Affiliation(s)
- Laure Aurelian
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
17
|
Li X, Xia R, Zhao J, Wang D, Mao Q, Xia H. A one-step ligation system for rapid generation of armed, conditionally-replicating adenoviruses. Biotechnol Lett 2013; 35:1215-21. [PMID: 23609233 DOI: 10.1007/s10529-013-1214-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 04/08/2013] [Indexed: 11/29/2022]
Abstract
Conditionally-replicating adenoviruses (CRAd) are becoming a promising strategy for cancer therapy. However, the generation of CRAd is complicated and time-consuming, especially for armed CRAd Ad5-D24. A one-step ligation system is now reported for rapid construction of an armed CRAd Ad5-D24 carrying two foreign transgenes. The system has the following features: (1) the CRAd backbone pCRAd5-D24-BS-Lacza contains two unique restriction sites between the fiber and E4 for easy introduction of a transgene expression cassette; and (2) a bi-directional promoter in pshuttle-BS-Dual allows for the expression of two independent transgenes. As proof of principle, Ad5-D24 CRAd carrying eGFP and mCherry was generated and examined. This novel ligation system will be a versatile and efficient tool in cancer viral gene therapy.
Collapse
Affiliation(s)
- Xing Li
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an, 710062, People's Republic of China
| | | | | | | | | | | |
Collapse
|
18
|
Angarita FA, Acuna SA, Ottolino-Perry K, Zerhouni S, McCart JA. Mounting a strategic offense: fighting tumor vasculature with oncolytic viruses. Trends Mol Med 2013; 19:378-92. [PMID: 23540715 DOI: 10.1016/j.molmed.2013.02.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 02/23/2013] [Accepted: 02/27/2013] [Indexed: 10/27/2022]
Abstract
Blood supply within a tumor drives progression and ultimately allows for metastasis. Many anticancer therapies target tumor vasculature, but their individual effectiveness is limited because they induce indirect cell death. Agents that disrupt nascent and/or established tumor vasculature while simultaneously killing cancer cells would certainly have a greater impact. Oncolytic virotherapy utilizes attenuated viruses that replicate specifically within a tumor. They induce cytotoxicity through a combination of direct cell lysis, antitumor immune stimulation, and recently identified antitumor vascular effects. This review summarizes the novel preclinical and clinical evidence regarding the antitumor vascular effects of oncolytic viruses, which include infection and lysis of tumor endothelial cells, natural or genetically engineered antiangiogenic properties, and combination therapy with clinically approved antivascular agents.
Collapse
Affiliation(s)
- Fernando A Angarita
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, Toronto, Ontario, M5G 2M1 Canada
| | | | | | | | | |
Collapse
|