1
|
Tang M, Ma G, Xu C, Yang H, Lin H, Bian C, Hu C, Lu M, Chen L, Jie W, Yue Z, Jian J, Sun Y, Yan H, Zhou J, Zhang X, Liao S, Li Z, Cai S, Wu Y, Yang K, Xiong Y, Zhao Y, Lv Z, Xu X, Liu C, Xin P, Ye L, Cui X, Shi Q, Chen X, Xu R. A facultative plasminogen-independent thrombolytic enzyme from Sipunculus nudus. Nat Commun 2025; 16:3852. [PMID: 40274794 PMCID: PMC12022309 DOI: 10.1038/s41467-025-58915-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: 06/27/2023] [Accepted: 04/04/2025] [Indexed: 04/26/2025] Open
Abstract
Current thrombolytic therapies primarily function by converting plasminogen into plasmin, a process dependent on the fibrin-activator complex. This dependence, coupled with the substantial molecular size of plasmin, constrains its effectiveness in degrading D-dimer and restricts its diffusion within thrombi. Here, we introduce a small facultative plasminogen-independent thrombolytic enzyme, snFPITE, isolated from Sipunculus nudus. Compared to traditional thrombolytic agents, snFPITE does not require plasminogen for thrombolysis, although its presence enhances lytic activity. This enzyme fully degrades cross-linked fibrin without leaving residual nondegradable D-dimer and generates a smaller fibrinolytic-active agent from plasminogen. A series of male rats and mice models further confirm that snFPITE is a safety injectable thrombolytic agent. Mechanistically, snFPITE activates plasminogen and degrades fibrin(ogen) in a multisite cleavage manner. snFPITE is inhibited by plasminogen activator inhibitor 1 and α2-antiplasmin via a competitive inhibition. We further identify 28 snFPITE candidate sequences, of which 10 are confirmed as functional genes.
Collapse
Affiliation(s)
- Mingqing Tang
- Engineering Research Centre of Molecular Medicine of Ministry of Education, Fujian Key Laboratory of Molecular Medicine, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, Xiamen Key Laboratory of Marine and Gene Drugs, School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China
| | - Guoxing Ma
- Engineering Research Centre of Molecular Medicine of Ministry of Education, Fujian Key Laboratory of Molecular Medicine, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, Xiamen Key Laboratory of Marine and Gene Drugs, School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China
- Department of Life Sciences, Tangshan Normal University, Tangshan, Hebei, China
| | | | - Hui Yang
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, Shanxi, China
| | - Hongjun Lin
- Engineering Research Centre of Molecular Medicine of Ministry of Education, Fujian Key Laboratory of Molecular Medicine, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, Xiamen Key Laboratory of Marine and Gene Drugs, School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China
| | - Chao Bian
- Laboratory of Aquatic Genomics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
| | - Chengjia Hu
- Engineering Research Centre of Molecular Medicine of Ministry of Education, Fujian Key Laboratory of Molecular Medicine, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, Xiamen Key Laboratory of Marine and Gene Drugs, School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China
| | - Meiling Lu
- Engineering Research Centre of Molecular Medicine of Ministry of Education, Fujian Key Laboratory of Molecular Medicine, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, Xiamen Key Laboratory of Marine and Gene Drugs, School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China
| | - Lei Chen
- Engineering Research Centre of Molecular Medicine of Ministry of Education, Fujian Key Laboratory of Molecular Medicine, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, Xiamen Key Laboratory of Marine and Gene Drugs, School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China
| | - Wencai Jie
- BGI Genomics, Shenzhen, Guangdong, China
| | - Zhen Yue
- BGI Research, Sanya, Hainan, China
| | | | - Yuqing Sun
- Engineering Research Centre of Molecular Medicine of Ministry of Education, Fujian Key Laboratory of Molecular Medicine, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, Xiamen Key Laboratory of Marine and Gene Drugs, School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China
| | - Hui Yan
- Engineering Research Centre of Molecular Medicine of Ministry of Education, Fujian Key Laboratory of Molecular Medicine, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, Xiamen Key Laboratory of Marine and Gene Drugs, School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China
| | - Jingjing Zhou
- Engineering Research Centre of Molecular Medicine of Ministry of Education, Fujian Key Laboratory of Molecular Medicine, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, Xiamen Key Laboratory of Marine and Gene Drugs, School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China
| | - Xianying Zhang
- Engineering Research Centre of Molecular Medicine of Ministry of Education, Fujian Key Laboratory of Molecular Medicine, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, Xiamen Key Laboratory of Marine and Gene Drugs, School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China
| | - Shengye Liao
- Engineering Research Centre of Molecular Medicine of Ministry of Education, Fujian Key Laboratory of Molecular Medicine, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, Xiamen Key Laboratory of Marine and Gene Drugs, School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China
| | - Zhaofa Li
- Engineering Research Centre of Molecular Medicine of Ministry of Education, Fujian Key Laboratory of Molecular Medicine, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, Xiamen Key Laboratory of Marine and Gene Drugs, School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China
| | - Shuangfeng Cai
- Engineering Research Centre of Molecular Medicine of Ministry of Education, Fujian Key Laboratory of Molecular Medicine, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, Xiamen Key Laboratory of Marine and Gene Drugs, School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China
| | - Yaqing Wu
- Engineering Research Centre of Molecular Medicine of Ministry of Education, Fujian Key Laboratory of Molecular Medicine, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, Xiamen Key Laboratory of Marine and Gene Drugs, School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China
| | - Kexin Yang
- Engineering Research Centre of Molecular Medicine of Ministry of Education, Fujian Key Laboratory of Molecular Medicine, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, Xiamen Key Laboratory of Marine and Gene Drugs, School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China
| | - Yanan Xiong
- Engineering Research Centre of Molecular Medicine of Ministry of Education, Fujian Key Laboratory of Molecular Medicine, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, Xiamen Key Laboratory of Marine and Gene Drugs, School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China
| | - Yonggang Zhao
- Engineering Research Centre of Molecular Medicine of Ministry of Education, Fujian Key Laboratory of Molecular Medicine, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, Xiamen Key Laboratory of Marine and Gene Drugs, School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China
| | - Zhimin Lv
- Engineering Research Centre of Molecular Medicine of Ministry of Education, Fujian Key Laboratory of Molecular Medicine, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, Xiamen Key Laboratory of Marine and Gene Drugs, School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China
- Xiamen Institute of Medicine and Technology, Xiamen, Fujian, China
| | - Xiaoming Xu
- Engineering Research Centre of Molecular Medicine of Ministry of Education, Fujian Key Laboratory of Molecular Medicine, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, Xiamen Key Laboratory of Marine and Gene Drugs, School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China
- Xiamen Institute of Medicine and Technology, Xiamen, Fujian, China
| | - Chuang Liu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Pengliang Xin
- Department of Haematology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian, China
| | - Lichao Ye
- Department of Neurology, The Second Affiliated Hospital, The Second Clinical Medical College, Fujian Medical University, Quanzhou, Fujian, China
| | - Xiuling Cui
- Engineering Research Centre of Molecular Medicine of Ministry of Education, Fujian Key Laboratory of Molecular Medicine, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, Xiamen Key Laboratory of Marine and Gene Drugs, School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China.
| | - Qiong Shi
- Laboratory of Aquatic Genomics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China.
| | - Xi Chen
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, Shanxi, China.
| | - Ruian Xu
- Engineering Research Centre of Molecular Medicine of Ministry of Education, Fujian Key Laboratory of Molecular Medicine, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, Xiamen Key Laboratory of Marine and Gene Drugs, School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China.
- Xiamen Institute of Medicine and Technology, Xiamen, Fujian, China.
| |
Collapse
|
2
|
Xue T, Wang X, Ru J, Zhang L, Yin H. The inhibitory effect of human umbilical cord mesenchymal stem cells expressing anti-HAAH scFv-sTRAIL fusion protein on glioma. Front Bioeng Biotechnol 2022; 10:997799. [DOI: 10.3389/fbioe.2022.997799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 10/26/2022] [Indexed: 11/10/2022] Open
Abstract
Glioma is the most common malignant intracranial tumor with low 5-year survival rate. In this study, we constructed a plasmid expressing anti-HAAH single-chain antibody and sTRAIL fusion protein (scFv-sTRAIL), and explored the effects of the double gene modified human umbilical cord mesenchyreal stem cells (hucMSCs) on the growth of glioma in vitro and in vivo. The isolated hucMSCs were identified by detecting the adipogenic differentiation ability and the osteogenic differentiation ability. The phenotypes of hucMSCs were determined by the flow cytometry. The hucMSCs were infected with lentivirus expression scFv-sTRAIL fusion protein. The expression of sTRAIL in hucMSCs were detected by immunofluorescence staining, western blot and ELISA. The tropism of hucMSCs toward U87G cells was assessed by transwell assay. The inhibitory effect of hucMSCs on U87G cells were explored by CCK8 and apoptosis assay. The xenograft tumor was established by subcutaneously injection of U87G cells into the back of mice. The hucMSCs were injected via tail veins. The inhibitory effect of hucMSCs on glioma in vivo was assessed by TUNEL assay. The hucMSCs migrated into the xenograft tumor were revealed by detecting the green fluorescent. The results showed that the scFv-sTRAIL expression did not affect the phenotypes of hucMSCs. The scFv-sTRAIL expression promoted the tropism of hucMSCs toward U87G cells, enhanced the inhibitory effect and tumor killing effect of hucMSCs on U87G cells. The in vivo study showed that hucMSCs expressing scFv-sTRAIL demonstrated significantly higher inhibitory effect and tumor killing effect than hucMSCs expressing sTRAIL. The green fluorescence intensity in the mice injected with hucMSCs expressing scFv-sTRAIL was significantly higher than that injected with hucMSCs expressing sTRAIL. These data suggested that the scFv conferred the targeting effect of hucMSCs tropism towards the xenograft tumor. In conclusion, the hucMSCs expressing scFv-sTRAIL fusion protein gained the capability to target and kill gliomas cells in vitro and in vivo. These findings shed light on a potential therapy for glioma treatment.
Collapse
|
3
|
Meumann N, Schmithals C, Elenschneider L, Hansen T, Balakrishnan A, Hu Q, Hook S, Schmitz J, Bräsen JH, Franke AC, Olarewaju O, Brandenberger C, Talbot SR, Fangmann J, Hacker UT, Odenthal M, Ott M, Piiper A, Büning H. Hepatocellular Carcinoma Is a Natural Target for Adeno-Associated Virus (AAV) 2 Vectors. Cancers (Basel) 2022; 14:cancers14020427. [PMID: 35053588 PMCID: PMC8774135 DOI: 10.3390/cancers14020427] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/21/2021] [Accepted: 01/11/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Gene therapy is a novel approach to treat diseases by introducing corrective genetic information into target cells. Adeno-associated virus vectors are the most frequently applied gene delivery tools for in vivo gene therapy and are also studied as part of innovative anticancer strategies. Here, we report on the natural preference of AAV2 vectors for hepatocellular carcinoma (HCC) compared to nonmalignant liver cells in mice and human tissue. This preference in transduction is due to the improved intracellular processing of AAV2 vectors in HCC, resulting in significantly more vector genomes serving as templates for transcription in the cell nucleus. Based on this natural tropism for HCC, novel therapeutic strategies can be designed or existing therapeutic approaches can be strengthened as they currently result in only a minor improvement of the poor prognosis for most liver cancer patients. Abstract Although therapeutic options are gradually improving, the overall prognosis for patients with hepatocellular carcinoma (HCC) is still poor. Gene therapy-based strategies are developed to complement the therapeutic armamentarium, both in early and late-stage disease. For efficient delivery of transgenes with antitumor activity, vectors demonstrating preferred tumor tropism are required. Here, we report on the natural tropism of adeno-associated virus (AAV) serotype 2 vectors for HCC. When applied intravenously in transgenic HCC mouse models, similar amounts of vectors were detected in the liver and liver tumor tissue. In contrast, transduction efficiency, as indicated by the level of transgene product, was moderate in the liver but was elevated up to 19-fold in mouse tumor tissue. Preferred transduction of HCC compared to hepatocytes was confirmed in precision-cut liver slices from human patient samples. Our mechanistic studies revealed that this preference is due to the improved intracellular processing of AAV2 vectors in HCC, resulting, for example, in nearly 4-fold more AAV vector episomes that serve as templates for gene transcription. Given this background, AAV2 vectors ought to be considered to strengthen current—or develop novel—strategies for treating HCC.
Collapse
Affiliation(s)
- Nadja Meumann
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (N.M.); (A.-C.F.); (O.O.); (U.T.H.)
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany;
| | - Christian Schmithals
- Department of Medicine 1, University Hospital, Goethe University Frankfurt, 60590 Frankfurt, Germany; (C.S.); (A.P.)
| | - Leroy Elenschneider
- Fraunhofer Institute for Toxicology and Experimental Medicine Preclinical Pharmacology and In-Vitro Toxicology, 30625 Hannover, Germany; (L.E.); (T.H.)
| | - Tanja Hansen
- Fraunhofer Institute for Toxicology and Experimental Medicine Preclinical Pharmacology and In-Vitro Toxicology, 30625 Hannover, Germany; (L.E.); (T.H.)
| | - Asha Balakrishnan
- Clinic for Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany; (A.B.); (Q.H.); (S.H.); (M.O.)
- Twincore Centre for Experimental and Clinical Infection Research, 30625 Hannover, Germany
| | - Qingluan Hu
- Clinic for Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany; (A.B.); (Q.H.); (S.H.); (M.O.)
- Twincore Centre for Experimental and Clinical Infection Research, 30625 Hannover, Germany
| | - Sebastian Hook
- Clinic for Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany; (A.B.); (Q.H.); (S.H.); (M.O.)
- Twincore Centre for Experimental and Clinical Infection Research, 30625 Hannover, Germany
| | - Jessica Schmitz
- Nephropathology Unit, Institute of Pathology, Hannover Medical School, 30625 Hannover, Germany; (J.S.); (J.H.B.)
| | - Jan Hinrich Bräsen
- Nephropathology Unit, Institute of Pathology, Hannover Medical School, 30625 Hannover, Germany; (J.S.); (J.H.B.)
| | - Ann-Christin Franke
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (N.M.); (A.-C.F.); (O.O.); (U.T.H.)
| | - Olaniyi Olarewaju
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (N.M.); (A.-C.F.); (O.O.); (U.T.H.)
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Christina Brandenberger
- Institute of Functional and Applied Anatomy, Hannover Medical School, 30625 Hannover, Germany;
- Biomedical Research in Endstage and Obstructive Lung Research (BREATH), German Center for Lung Research (DZL), 30625 Hannover, Germany
| | - Steven R. Talbot
- Institute for Laboratory Animal Science, Hannover Medical School, 30625 Hannover, Germany;
| | - Josef Fangmann
- KRH Klinikum Siloah, Liver Center Hannover (LCH), 30459 Hannover, Germany;
| | - Ulrich T. Hacker
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (N.M.); (A.-C.F.); (O.O.); (U.T.H.)
- Department of Oncology, Gastroenterology, Hepatology, Pulmonology, and Infectious Diseases, University Cancer Center Leipzig (UCCL), Leipzig University Medical Center, 04103 Leipzig, Germany
| | - Margarete Odenthal
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany;
- Institute of Pathology, University Hospital Cologne, 50931 Cologne, Germany
| | - Michael Ott
- Clinic for Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany; (A.B.); (Q.H.); (S.H.); (M.O.)
- Twincore Centre for Experimental and Clinical Infection Research, 30625 Hannover, Germany
| | - Albrecht Piiper
- Department of Medicine 1, University Hospital, Goethe University Frankfurt, 60590 Frankfurt, Germany; (C.S.); (A.P.)
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Hildegard Büning
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (N.M.); (A.-C.F.); (O.O.); (U.T.H.)
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany;
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany
- Correspondence: ; Tel.: +49-511-532-5106
| |
Collapse
|
4
|
Younis MA, Khalil IA, Harashima H. Gene Therapy for Hepatocellular Carcinoma: Highlighting the Journey from Theory to Clinical Applications. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000087] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Mahmoud A. Younis
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences Hokkaido University Kita‐12, Nishi‐6, Kita‐ku Sapporo 060‐0812 Japan
- Faculty of Pharmacy Assiut University Assiut 71526 Egypt
| | - Ikramy A. Khalil
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences Hokkaido University Kita‐12, Nishi‐6, Kita‐ku Sapporo 060‐0812 Japan
- Faculty of Pharmacy Assiut University Assiut 71526 Egypt
| | - Hideyoshi Harashima
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences Hokkaido University Kita‐12, Nishi‐6, Kita‐ku Sapporo 060‐0812 Japan
| |
Collapse
|
5
|
Hacker UT, Bentler M, Kaniowska D, Morgan M, Büning H. Towards Clinical Implementation of Adeno-Associated Virus (AAV) Vectors for Cancer Gene Therapy: Current Status and Future Perspectives. Cancers (Basel) 2020; 12:E1889. [PMID: 32674264 PMCID: PMC7409174 DOI: 10.3390/cancers12071889] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 02/06/2023] Open
Abstract
Adeno-associated virus (AAV) vectors have gained tremendous attention as in vivo delivery systems in gene therapy for inherited monogenetic diseases. First market approvals, excellent safety data, availability of large-scale production protocols, and the possibility to tailor the vector towards optimized and cell-type specific gene transfer offers to move from (ultra) rare to common diseases. Cancer, a major health burden for which novel therapeutic options are urgently needed, represents such a target. We here provide an up-to-date overview of the strategies which are currently developed for the use of AAV vectors in cancer gene therapy and discuss the perspectives for the future translation of these pre-clinical approaches into the clinic.
Collapse
Affiliation(s)
- Ulrich T. Hacker
- Department of Oncology, Gastroenterology, Hepatology, Pulmonology, and Infectious Diseases, University Cancer Center Leipzig (UCCL), Leipzig University Medical Center, 04103 Leipzig, Germany;
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (M.B.); (M.M.)
| | - Martin Bentler
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (M.B.); (M.M.)
| | - Dorota Kaniowska
- Department of Oncology, Gastroenterology, Hepatology, Pulmonology, and Infectious Diseases, University Cancer Center Leipzig (UCCL), Leipzig University Medical Center, 04103 Leipzig, Germany;
| | - Michael Morgan
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (M.B.); (M.M.)
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Hildegard Büning
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (M.B.); (M.M.)
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Inhoffenstraße 7, 38124 Braunschweig, Germany
| |
Collapse
|
6
|
Recombinant Viruses for Cancer Therapy. Biomedicines 2018; 6:biomedicines6040094. [PMID: 30257488 PMCID: PMC6316473 DOI: 10.3390/biomedicines6040094] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 09/17/2018] [Accepted: 09/20/2018] [Indexed: 12/28/2022] Open
Abstract
Recombinant viruses are novel therapeutic agents that can be utilized for treatment of various diseases, including cancers. Recombinant viruses can be engineered to express foreign transgenes and have a broad tropism allowing gene expression in a wide range of host cells. They can be selected or designed for specific therapeutic goals; for example, recombinant viruses could be used to stimulate host immune response against tumor-specific antigens and therefore overcome the ability of the tumor to evade the host's immune surveillance. Alternatively, recombinant viruses could express immunomodulatory genes which stimulate an anti-cancer immune response. Oncolytic viruses can replicate specifically in tumor cells and induce toxic effects leading to cell lysis and apoptosis. However, each of these approaches face certain difficulties that must be resolved to achieve maximum therapeutic efficacy. In this review we discuss actively developing approaches for cancer therapy based on recombinant viruses, problems that need to be overcome, and possible prospects for further development of recombinant virus based therapy.
Collapse
|
7
|
Dhungel B, Jayachandran A, Layton CJ, Steel JC. Seek and destroy: targeted adeno-associated viruses for gene delivery to hepatocellular carcinoma. Drug Deliv 2017; 24:289-299. [PMID: 28165834 PMCID: PMC8241004 DOI: 10.1080/10717544.2016.1247926] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common form of primary liver cancer with high incidence globally. Increasing mortality and morbidity rates combined with limited treatment options available for advanced HCC press for novel and effective treatment modalities. Gene therapy represents one of the most promising therapeutic options. With the recent approval of herpes simplex virus for advanced melanoma, the field of gene therapy has received a major boost. Adeno-associated virus (AAV) is among the most widely used and effective viral vectors today with safety and efficacy demonstrated in a number of human clinical trials. This review identifies the obstacles for effective AAV based gene delivery to HCC which primarily include host immune responses and off-target effects. These drawbacks could be more pronounced for HCC because of the underlying liver dysfunction in most of the patients. We discuss approaches that could be adopted to tackle these shortcomings and manufacture HCC-targeted vectors. The combination of transductional targeting by modifying the vector capsid and transcriptional targeting using HCC-specific promoters has the potential to produce vectors which can specifically seek HCC and deliver therapeutic gene without significant side effects. Finally, the identification of novel HCC-specific ligands and promoters should facilitate and expedite this process.
Collapse
Affiliation(s)
- Bijay Dhungel
- a Gallipoli Medical Research Institute, Greenslopes Private Hospital , Brisbane , QLD , Australia.,b School of Medicine, The University of Queensland , Brisbane , QLD , Australia.,c University of Queensland Diamantina Institute, Translational Research Institute , Woolloongabba , QLD , Australia , and
| | - Aparna Jayachandran
- a Gallipoli Medical Research Institute, Greenslopes Private Hospital , Brisbane , QLD , Australia.,b School of Medicine, The University of Queensland , Brisbane , QLD , Australia
| | - Christopher J Layton
- b School of Medicine, The University of Queensland , Brisbane , QLD , Australia.,d Ophthalmology Department, Gallipoli Medical Research Institute, Greenslopes Private Hospital , Brisbane , QLD , Australia
| | - Jason C Steel
- a Gallipoli Medical Research Institute, Greenslopes Private Hospital , Brisbane , QLD , Australia.,b School of Medicine, The University of Queensland , Brisbane , QLD , Australia
| |
Collapse
|
8
|
Ru Q, Li W, Wang X, Zhang S, Chen L, Zhang Y, Ge Y, Zu Y, Liu Y, Zheng D. Preclinical study of rAAV2-sTRAIL: pharmaceutical efficacy, biodistribution and safety in animals. Cancer Gene Ther 2017; 24:251-258. [PMID: 28429751 DOI: 10.1038/cgt.2017.12] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Revised: 02/02/2017] [Accepted: 03/07/2017] [Indexed: 12/31/2022]
Abstract
The recombinant sTRAIL has been in clinical trial for various human malignancies. However, the half-life time of sTRAIL is very short, which might be an important factor influencing its clinical efficacy for cancer therapy. We previously reported the recombinant adeno-associated virus (AAV)-encoding sTRAIL95-281-mediated sTRAIL expression in vivo up to 8 months and suppressed tumor growth markedly in mouse xenografts. In the present study, we further evaluated the clinical potency for cancer gene therapy and the safety in mouse and non-human primates. The mouse models with HCT-116, NCI-H460 and BEL-7402 cancers were injected intraperitoneally with a single dose of 1.0 × 1011, 1.0 × 1010 and 1.0 × 109 vg of rAAV2-sTRAIL95-281 virus, respectively. The cynomolgus monkeys were injected (i.m.) with a single dose of rAAV2-sTRAIL95-281 of 1 × 1011, 3 × 1011 and 1 × 1012 vg, corresponding to 6-, 20- and 60-fold of intended use dosage for humans, respectively. The efficacy, pharmacology and toxicity of rAAV-sTRAIL in the animals were analyzed accordingly. The tumor inhibitory rates reached 44-76%, 48-52% and 55-74% in the three tumor models, respectively, and they had no influence on mouse spontaneous activity. Administration (s.c.) of a single dose of rAAV2-sTRAIL95-281 virus of 1.0 × 109 or 1.0 × 1010 vg in mice with implanted tumor led to mainly distribution in the spleen, liver, implanted tumor, blood, injected site of muscle and bone marrow. Two weeks later, there was no rAAV2-sTRAIL95-281 detected in blood and bone marrow, and it significantly decreased in other tissues and organs and then gradually cleared away in 4-12 weeks after administration. There was no rAAV2-sTRAIL accumulation in the animal's body and no influence on the body weights. Administration (i.v.) did not cause animal death, and no dose-related abnormal clinical symptoms were found in the mice. There were no abnormal tissue and organ found in all animals. Long-term toxicity test in cynomolgus monkeys did not cause rAAV2-sTRAIL95-281-related toxic and side effects, except that anti-AAV and anti-sTRAIL antibodies were generated. In conclusion, these data demonstrated that administration of rAAV2-sTRAIL95-281 in mice and in cynomolgus monkeys is safe without obvious toxic and side effects to the animals, and throw light on pharmacokinetics and safety in human clinical trials for cancer gene therapy.
Collapse
Affiliation(s)
- Q Ru
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - W Li
- National Institutes for Food and Drug Control, National Center for Safety Evaluation of Drugs, Beijing, China
| | - X Wang
- National Institutes for Food and Drug Control, National Center for Safety Evaluation of Drugs, Beijing, China
| | - S Zhang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences &Peking Union Medical College, Beijing, China
| | - L Chen
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Y Zhang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Y Ge
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Y Zu
- Obio Technology (Shanghai) Corp. Ltd, Shanghai, China
| | - Y Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - D Zheng
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
9
|
Santiago-Ortiz JL, Schaffer DV. Adeno-associated virus (AAV) vectors in cancer gene therapy. J Control Release 2016; 240:287-301. [PMID: 26796040 PMCID: PMC4940329 DOI: 10.1016/j.jconrel.2016.01.001] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 12/08/2015] [Accepted: 01/02/2016] [Indexed: 02/06/2023]
Abstract
Gene delivery vectors based on adeno-associated virus (AAV) have been utilized in a large number of gene therapy clinical trials, which have demonstrated their strong safety profile and increasingly their therapeutic efficacy for treating monogenic diseases. For cancer applications, AAV vectors have been harnessed for delivery of an extensive repertoire of transgenes to preclinical models and, more recently, clinical trials involving certain cancers. This review describes the applications of AAV vectors to cancer models and presents developments in vector engineering and payload design aimed at tailoring AAV vectors for transduction and treatment of cancer cells. We also discuss the current status of AAV clinical development in oncology and future directions for AAV in this field.
Collapse
Affiliation(s)
- Jorge L Santiago-Ortiz
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA
| | - David V Schaffer
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA; Department of Bioengineering, University of California, Berkeley, CA, USA; Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA; The Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA.
| |
Collapse
|
10
|
Systemically administered AAV9-sTRAIL combats invasive glioblastoma in a patient-derived orthotopic xenograft model. MOLECULAR THERAPY-ONCOLYTICS 2016; 3:16017. [PMID: 27382645 PMCID: PMC4916948 DOI: 10.1038/mto.2016.17] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 05/06/2016] [Indexed: 12/22/2022]
Abstract
Adeno-associated virus (AAV) vectors expressing tumoricidal genes injected directly into brain tumors have shown some promise, however, invasive tumor cells are relatively unaffected. Systemic injection of AAV9 vectors provides widespread delivery to the brain and potentially the tumor/microenvironment. Here we assessed AAV9 for potential glioblastoma therapy using two different promoters driving the expression of the secreted anti-cancer agent sTRAIL as a transgene model; the ubiquitously active chicken β-actin (CBA) promoter and the neuron-specific enolase (NSE) promoter to restrict expression in brain. Intravenous injection of AAV9 vectors encoding a bioluminescent reporter showed similar distribution patterns, although the NSE promoter yielded 100-fold lower expression in the abdomen (liver), with the brain-to-liver expression ratio remaining the same. The main cell types targeted by the CBA promoter were astrocytes, neurons and endothelial cells, while expression by NSE promoter mostly occurred in neurons. Intravenous administration of either AAV9-CBA-sTRAIL or AAV9-NSE-sTRAIL vectors to mice bearing intracranial patient-derived glioblastoma xenografts led to a slower tumor growth and significantly increased survival, with the CBA promoter having higher efficacy. To our knowledge, this is the first report showing the potential of systemic injection of AAV9 vector encoding a therapeutic gene for the treatment of brain tumors.
Collapse
|
11
|
Genetic Manipulation of Brown Fat Via Oral Administration of an Engineered Recombinant Adeno-associated Viral Serotype Vector. Mol Ther 2016; 24:1062-1069. [PMID: 26857843 DOI: 10.1038/mt.2016.34] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 01/27/2016] [Indexed: 12/24/2022] Open
Abstract
Recombinant adeno-associated virus (rAAV) vectors are attractive vehicles for gene therapy. Gene delivery to the adipose tissue using naturally occurring AAV serotypes is less successful compared to liver and muscle. Here, we demonstrate that oral administration of an engineered serotype Rec2 led to preferential transduction of brown fat with absence of transduction in the gastrointestinal track. Among the six natural and engineered serotypes being compared, Rec2 was the most efficient serotype achieving high level transduction at a dose 1~2 orders lower than reported doses for systemic administration. Overexpressing vascular endothelial growth factor (VEGF) in brown fat via oral administration of Rec2-VEGF vector increased the brown fat mass and enhanced thermogenesis. In contrast, knockdown VEGF in brown fat of VEGF (loxP) mice via Rec2-Cre vector hampered cold response and decreased brown fat mass. Oral administration of Rec2 vector provides a novel tool to genetically manipulate brown fat for research and therapeutic applications.
Collapse
|
12
|
Gao J, Wang D, Liu D, Liu M, Ge Y, Jiang M, Liu Y, Zheng D. Tumor necrosis factor-related apoptosis-inducing ligand induces the expression of proinflammatory cytokines in macrophages and re-educates tumor-associated macrophages to an antitumor phenotype. Mol Biol Cell 2015. [PMID: 26224317 PMCID: PMC4569310 DOI: 10.1091/mbc.e15-04-0209] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
This study reveals that tumor necrosis factor–related apoptosis-inducing ligand (TRAIL) induces expression of IL-1β, IL-6, and tumor necrosis factor α in macrophages, especially in tumor-associated macrophages (TAMs). TRAIL re-educates TAMs to an M1-like phenotype and induces their cytotoxicity to tumor cells. This study provides new evidence for TRAIL in immune regulation of macrophages and sheds light on TRAIL-based antitumor therapy in human patients. Tumor necrosis factor–related apoptosis-inducing ligand (TRAIL) is a promising candidate for cancer therapy, because it can induce apoptosis in various tumor cells but not in most normal cells. Although it is well known that TRAIL and its receptors are expressed in many types of normal cells, including immune cells, their immunological effects and regulatory mechanisms are still obscure. In the present study, we demonstrated that TRAIL affected the activity of NF-κB (nuclear factor-κB) and the expression of its downstream proinflammatory cytokines IL-1β (interleukin-1β), IL-6, and tumor necrosis factor α in macrophages. TRAIL also induced microRNA-146a (miR-146a) expression in an NF-κB–dependent manner. As a result, miR-146a was involved as a negative-feedback regulator in the down-regulation of proinflammatory cytokine expression. In addition, the suppression of histone deacetylase (HDAC) activities by trichostatin A improved miR-146a expression due to the up-regulation of the DNA-binding activity of NF-κB at the miR-146a promoter in TRAIL-induced macrophages, suggesting that histone acetylation was involved in the suppression of miR-146a expression. Further investigation revealed that the HDAC subtype HDAC1 directly regulated the expression of miR-146a in TRAIL-stimulated macrophages. Finally, the TRAIL-sensitive human non small cell lung carcinoma cell line NCI-H460 was used to elucidate the physiological significance of TRAIL with respect to tumor-associated macrophages (TAMs). We demonstrated that TRAIL re-educated TAMs to an M1-like phenotype and induced cytotoxic effects in the tumor cells. These data provide new evidence for TRAIL in the immune regulation of macrophages and may shed light on TRAIL-based antitumor therapy in human patients.
Collapse
Affiliation(s)
- Jing Gao
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Dongsheng Wang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Dan Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Min Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Yehua Ge
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Minghong Jiang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Yanxin Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Dexian Zheng
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| |
Collapse
|
13
|
Luo J, Luo Y, Sun J, Zhou Y, Zhang Y, Yang X. Adeno-associated virus-mediated cancer gene therapy: current status. Cancer Lett 2014; 356:347-56. [PMID: 25444906 DOI: 10.1016/j.canlet.2014.10.045] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 10/30/2014] [Accepted: 10/31/2014] [Indexed: 01/18/2023]
Abstract
Gene therapy is one of the frontiers of modern medicine. Adeno-associated virus (AAV)-mediated gene therapy is becoming a promising approach to treat a variety of diseases and cancers. AAV-mediated cancer gene therapies have rapidly advanced due to their superiority to other gene-carrying vectors, such as the lack of pathogenicity, the ability to transfect both dividing and non-dividing cells, low host immune response, and long-term expression. This article reviews and provides up to date knowledge on AAV-mediated cancer gene therapy.
Collapse
Affiliation(s)
- Jingfeng Luo
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Qingchun Road NO.3, Hangzhou, Zhejiang, China
| | - Yuxuan Luo
- Department of Nephrology, Zhuji People's Hospital, Zhuji, Zhejiang, China
| | - Jihong Sun
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Qingchun Road NO.3, Hangzhou, Zhejiang, China
| | - Yurong Zhou
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Qingchun Road NO.3, Hangzhou, Zhejiang, China
| | - Yajing Zhang
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Qingchun Road NO.3, Hangzhou, Zhejiang, China
| | - Xiaoming Yang
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Qingchun Road NO.3, Hangzhou, Zhejiang, China; Image-Guided Bio-Molecular Intervention Research, Department of Radiology, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
14
|
Adeno-associated virus-mediated expression of recombinant CBD–HepII polypeptide of human fibronectin inhibits metastasis of breast cancer. Breast Cancer Res Treat 2013; 143:33-45. [DOI: 10.1007/s10549-013-2783-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 11/20/2013] [Indexed: 01/09/2023]
|
15
|
Hao ZM, Cai M, Lv YF, Huang YH, Li HH. Oral administration of recombinant adeno-associated virus-mediated bone morphogenetic protein-7 suppresses CCl(4)-induced hepatic fibrosis in mice. Mol Ther 2012; 20:2043-51. [PMID: 22850680 DOI: 10.1038/mt.2012.148] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Fibrogenesis and hepatocyte degeneration are the main pathological processes in chronic liver diseases. Transforming growth factor-β1 (TGF-β1) is the key profibrotic cytokine in hepatic fibrosis. Bone morphogenetic protein-7 (BMP-7) is a potent antagonist of TGF-β1 and an antifibrotic factor. In this study, we generated a recombinant adeno-associated virus carrying BMP-7 (AAV-BMP-7) and tested its ability to suppress carbon tetrachloride (CCl(4))-induced hepatic fibrosis when orally administered to mice. Our results show that the ectopic expression of BMP-7 in gastrointestinal (GI) mucosa due to the AAV-BMP-7 administration led to the long-term elevation of serum BMP-7 concentrations and resulted in the drastic amelioration of CCl(4)-induced hepatic fibrosis in BALB/c mice. Immunostaining for α-smooth muscle actin (α-SMA) and desmin demonstrated that AAV-BMP-7 inhibited the activation of hepatic stellate cells (HSCs) in the fibrotic mouse liver. Moreover, the ectopic expression of BMP-7 promoted hepatocyte proliferation, as confirmed by an increase in the amount of proliferating cell nuclear antigen (PCNA)-positive hepatocytes in the mice that received AAV-BMP-7. Our results clearly indicate that BMP-7 is capable of inhibiting hepatic fibrosis and promoting hepatocyte regeneration. We suggest that oral AAV-BMP-7 could be developed into a safe, simple, and effective therapy for hepatic fibrosis.
Collapse
Affiliation(s)
- Zhi-Ming Hao
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China.
| | | | | | | | | |
Collapse
|
16
|
Zheng L, Weilun Z, Minghong J, Yaxi Z, Shilian L, Yanxin L, Dexian Z. Adeno-associated virus-mediated doxycycline-regulatable TRAIL expression suppresses growth of human breast carcinoma in nude mice. BMC Cancer 2012; 12:153. [PMID: 22530952 PMCID: PMC3404920 DOI: 10.1186/1471-2407-12-153] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Accepted: 04/24/2012] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) functions as a cytokine to selectively kill various cancer cells without toxicity to most normal cells. Numerous studies have demonstrated the potential use of recombinant soluble TRAIL as a cancer therapeutic agent. We have showed previous administration of a recombinant adeno-associated virus (rAAV) vector expressing soluble TRAIL results in an efficient suppression of human tumor growth in nude mice. In the present study, we introduced Tet-On gene expression system into the rAAV vector to control the soluble TRAIL expression and evaluate the efficiency of the system in cancer gene therapy. METHODS Controllability of the Tet-On system was determined by luciferase activity assay, and Western blotting and enzyme-linked immunoabsorbent assay. Cell viability was determined by MTT assay. The breast cancer xenograft animal model was established and recombinant virus was administrated through tail vein injection to evaluate the tumoricidal activity. RESULTS The expression of soluble TRAIL could be strictly controlled by the Tet-On system in both normal and cancer cells. Transduction of human cancer cell lines with rAAV-TRE-TRAIL&rAAV-Tet-On under the presence of inducer doxycycline resulted in a considerable cell death by apoptosis. Intravenous injection of the recombinant virus efficiently suppressed the growth of human breast carcinoma in nude mice when activated by doxycycline. CONCLUSION These data suggest that rAAV-mediated soluble TRAIL expression under the control of the Tet-On system is a promising strategy for breast cancer therapy.
Collapse
Affiliation(s)
- Liu Zheng
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005, China
| | - Zhang Weilun
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005, China
| | - Jiang Minghong
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005, China
| | - Zhang Yaxi
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005, China
| | - Liu Shilian
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005, China
| | - Liu Yanxin
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005, China
| | - Zheng Dexian
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005, China
| |
Collapse
|
17
|
Cheng B, Ling C, Dai Y, Lu Y, Glushakova LG, Gee SWY, McGoogan KE, Aslanidi GV, Park M, Stacpoole PW, Siemann D, Liu C, Srivastava A, Ling C. Development of optimized AAV3 serotype vectors: mechanism of high-efficiency transduction of human liver cancer cells. Gene Ther 2011; 19:375-84. [PMID: 21776025 PMCID: PMC3519243 DOI: 10.1038/gt.2011.105] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Our recent studies have revealed that among the 10 different commonly used adeno-associated virus (AAV) serotypes, AAV3 vectors transduce human liver cancer cells extremely efficiently because these cells express high levels of human hepatocyte growth factor receptor (hHGFR), and AAV3 utilizes hHGFR as a cellular co-receptor for viral entry. In this report, we provide further evidence that both extracellular as well as intracellular kinase domains of hHGFR are involved in AAV3 vector entry and AAV3-mediated transgene expression. We also document that AAV3 vectors are targeted for degradation by the host cell proteasome machinery, and that site-directed mutagenesis of surface-exposed tyrosine (Y) to phenylalanine (F) residues on AAV3 capsids significantly improves the transduction efficiency of Y701F, Y705F and Y731F mutant AAV3 vectors. The transduction efficiency of the Y705+731F double-mutant vector is significantly higher than each of the single mutants in liver cancer cells in vitro. In immunodeficient mouse xenograft models, direct intratumoral injection of AAV3 vectors also led to high-efficiency transduction of human liver tumor cells in vivo. We also document here that the optimized tyrosine-mutant AAV3 vectors lead to increased transduction efficiency following both intratumoral and tail-vein injections in vivo. The optimized tyrosine-mutant AAV3 serotype vectors containing proapoptotic genes should prove useful for the potential gene therapy of human liver cancers.
Collapse
Affiliation(s)
- B Cheng
- Division of Cellular and Molecular Therapy, Departments of Pediatics and Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, FL 32610-3633, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Norian LA, James BR, Griffith TS. Advances in Viral Vector-Based TRAIL Gene Therapy for Cancer. Cancers (Basel) 2011; 3:603-20. [PMID: 24212631 PMCID: PMC3756379 DOI: 10.3390/cancers3010603] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2010] [Revised: 01/28/2011] [Accepted: 01/30/2011] [Indexed: 12/16/2022] Open
Abstract
Numerous biologic approaches are being investigated as anti-cancer therapies in an attempt to induce tumor regression while circumventing the toxic side effects associated with standard chemo- or radiotherapies. Among these, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has shown particular promise in pre-clinical and early clinical trials, due to its preferential ability to induce apoptotic cell death in cancer cells and its minimal toxicity. One limitation of TRAIL use is the fact that many tumor types display an inherent resistance to TRAIL-induced apoptosis. To circumvent this problem, researchers have explored a number of strategies to optimize TRAIL delivery and to improve its efficacy via co-administration with other anti-cancer agents. In this review, we will focus on TRAIL-based gene therapy approaches for the treatment of malignancies. We will discuss the main viral vectors that are being used for TRAIL gene therapy and the strategies that are currently being attempted to improve the efficacy of TRAIL as an anti-cancer therapeutic.
Collapse
Affiliation(s)
- Lyse A. Norian
- Department of Urology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; E-Mail:
| | - Britnie R. James
- Interdisciplinary Graduate Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; E-Mail:
| | - Thomas S. Griffith
- Department of Urology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; E-Mail:
- Interdisciplinary Graduate Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; E-Mail:
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-319-335-7581; Fax: +1-319-353-4556
| |
Collapse
|
19
|
Jiang M, Liu Z, Xiang Y, Ma H, Liu S, Liu Y, Zheng D. Synergistic antitumor effect of AAV-mediated TRAIL expression combined with cisplatin on head and neck squamous cell carcinoma. BMC Cancer 2011; 11:54. [PMID: 21291526 PMCID: PMC3044652 DOI: 10.1186/1471-2407-11-54] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2010] [Accepted: 02/03/2011] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND Adeno-associated virus-2 (AAV-2)-mediated gene therapy is quite suitable for local or regional application in head and neck cancer squamous cell carcinoma (HNSCC). However, its low transduction efficiency has limited its further development as a therapeutic agent. DNA damaging agents have been shown to enhance AAV-mediated transgene expression. Cisplatin, one of the most effective chemotherapeutic agents, has been recognized to cause cancer cell death by apoptosis with a severe toxicity. This study aims to evaluate the role of cisplatin in AAV-mediated tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) expression and the effect on HNSCC both in vitro and in vivo. METHODS Five human HNSCC cell lines were treated with recombinant soluble TRAIL (rsTRAIL) and infected with AAV/TRAIL to estimate the sensitivity of the cancer cells to TRAIL-induced cytotoxicity. KB cells were infected with AAV/EGFP with or without cisplatin pretreatment to evaluate the effect of cisplatin on AAV-mediated gene expression. TRAIL expression was detected by ELISA and Western blot. Cytotoxicity was measured by MTT assay and Western blot analysis for caspase-3 and -8 activations. Following the in vitro experiments, TRAIL expression and its tumoricidal activity were analyzed in nude mice with subcutaneous xenografts of HNSCC. RESULTS HNSCC cell lines showed different sensitivities to rsTRAIL, and KB cells possessed both highest transduction efficacy of AAV and sensitivity to TRAIL among five cell lines. Preincubation of KB cells with subtherapeutic dosage of cisplatin significantly augmented AAV-mediated transgene expression in a heparin sulfate proteoglycan (HSPG)-dependent manner. Furthermore, cisplatin enhanced the killing efficacy of AAV/TRAIL by 3-fold on KB cell line. The AAV mediated TRAIL expression was observed in the xenografted tumors and significantly enhanced by cisplatin. AAV/TRAIL suppressed the tumors growth and cisplatin augmented the tumoricidal activity by two-fold. Furthermore, Combination treatment reduced cisplatin-caused body weight loss in nude mice. CONCLUSION The combination of AAV-mediated TRAIL gene expression and cisplatin had synergistic therapeutic effects on head and neck cancers and reduced the potential toxicity of cisplatin. These findings suggest that the combination of AAV/TRAIL and cisplatin may be a promising strategy for HNSCC therapy.
Collapse
Affiliation(s)
- Minghong Jiang
- National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Zheng Liu
- National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Yang Xiang
- National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Hong Ma
- National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Shilian Liu
- National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Yanxin Liu
- National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Dexian Zheng
- National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| |
Collapse
|
20
|
Suda T, Kamimura K, Kubota T, Tamura Y, Igarashi M, Kawai H, Aoyagi Y, Liu D. Progress toward liver-based gene therapy. Hepatol Res 2009; 39:325-340. [PMID: 19207594 DOI: 10.1111/j.1872-034x.2008.00479.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The liver is involved in the synthesis of serum proteins, regulation of metabolism and maintenance of homeostasis and provides a variety of opportunities for gene therapy. The enriched vasculature and blood circulation, fenestrated endothelium, abundant receptors on the plasma membranes of the liver cells, and effective transcription and translation machineries in the hepatocytes are some unique features that have been explored for delivery, and functional analysis, of genetic sequences in the liver. Both viral and non-viral methods have been developed for effective gene delivery and liver-based gene therapy. This review describes the fundamentals of gene delivery, and the preclinical and clinical progress that has been made toward gene therapy using the liver as a target.
Collapse
Affiliation(s)
- Takeshi Suda
- Division of Gastroenterology and Hepatology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Niigata, Japan
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Shepard BD, Badley AD. The Biology of TRAIL and the Role of TRAIL-Based Therapeutics in Infectious Diseases. ACTA ACUST UNITED AC 2009; 8:87-101. [PMID: 21857885 DOI: 10.2174/187152109787846060] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
TNF-related apoptosis inducing ligand (TRAIL) is a key mediator of the innate immune response to infection. While TRAIL-mediated apoptosis plays an essential role in the clearance of virus-infected cells, its physiologic role also includes immunosurveilance for cancer cells. Therapeutics that induce TRAIL-mediated apoptosis in cancer cells remain a focus of ongoing investigation in clinical trials, and much has been learned from these studies regarding the efficacy and toxicity of these interventions. These data, combined with data from numerous preclinical studies that detail the important and multifaceted role of TRAIL during infection with human immunodeficiency virus and other viruses, suggest that therapeutic exploitation of TRAIL signaling offers a novel and efficacious strategy for the management of infectious diseases.
Collapse
Affiliation(s)
- Brett D Shepard
- Mayo Clinic College of Medicine, Division of Infectious Diseases, Rochester, MN, 55905, USA
| | | |
Collapse
|
22
|
|
23
|
Wang Y, Huang F, Cai H, Zhong S, Liu X, Tan WS. Potent antitumor effect of TRAIL mediated by a novel adeno-associated viral vector targeting to telomerase activity for human hepatocellular carcinoma. J Gene Med 2008; 10:518-26. [PMID: 18338833 DOI: 10.1002/jgm.1177] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Adeno-associated virus (AAV) has rapidly become a promising gene delivery vehicle for its excellent advantages of low pathogenicity and long-term gene expression. However, lack of tissue specificity caused low efficiency of AAV transfer to target cells. The promoter of human telomerase reverse transcriptase (hTERT) has been implicated in mediating gene expression in cancer cells as hTERT is transcriptionally upregulated in most cancer cells. Thereby, the hTERT promoter becomes a good candidate to enhance the targeting efficiency of AAV in cancer cells. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) functions as a soluble cytokine to selectively kill various cancer cells without toxicity to most normal cells. It remains to be determined whether the hTERT promoter can efficiently mediate TRAIL gene therapy in cancer cells using AAV vector. METHODS A novel AAV vector containing the TRAIL gene under the control of the hTERT promoter (AAV-hTERT-TRAIL) was generated. The specific expression of hTERT-controlled genes was evaluated in cell lines. The antitumor efficacy of AAV-hTERT-TRAIL was assessed in tumor cell lines and human hepatocellular carcinoma xenograft mouse model. RESULTS TRAIL expression was observed in tumor cells infected with AAV-hTERT-TRAIL at both the protein and mRNA level. AAV-hTERT-TRAIL displayed cancer-specific cytotoxicity and induced tumor cell apoptosis. Moreover, in animal experiments, intratumoral administration of AAV-hTERT-TRAIL significantly suppressed the growth of xenograft tumors and resulted in tumor cell death. CONCLUSIONS AAVs in combination with hTERT-mediated therapeutic gene expression provide a promising targeting approach for developing effective therapy for human cancers. These data suggest that AAV-hTERT-TRAIL is a potent therapeutic agent for cancer therapy.
Collapse
Affiliation(s)
- Yigang Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | | | | | | | | | | |
Collapse
|
24
|
Zhang Y, Ma H, Zhang J, Liu S, Liu Y, Zheng D. AAV-mediated TRAIL gene expression driven by hTERT promoter suppressed human hepatocellular carcinoma growth in mice. Life Sci 2008; 82:1154-61. [PMID: 18485417 DOI: 10.1016/j.lfs.2008.03.023] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2007] [Revised: 03/10/2008] [Accepted: 03/19/2008] [Indexed: 12/14/2022]
Abstract
A major obstacle in the development of effective recombinant adeno-associated virus (rAAV) mediated gene therapy is infection specificity and gene targeting. In the present study, we investigated whether the human telomerase reverse transcriptase (hTERT) promoter could drive tumor-specific expression of the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), an apoptosis apoptosis-inducing protein with potential toxic effects on normal cells. Our data demonstrated that hTERT promoter-driven tumor-specific expression of TRAIL decreased the cellular viability of tumor cells, but not normal cells. TRAIL expression driven by hTERT promoter inhibited tumor growth significantly in vivo and combination of viral infection with 5-fluorouracil (5-Fu) suppressed tumor growth more efficiently. Intra-venous injection of virus showed that the recombinant virus was predominantly distributed in the liver, but not in other major tissues tested, and no transgene expression was detected in the liver. Furthermore, serum enzyme and liver histology analysis confirmed that liver function is unaffected by TRAIL expression, significant as the liver is frequently metastasized and scattered with tumors from other organs, which are unpractical to treat by intra-tumor injection. Together our results demonstrate that rAAV-mediated TRAIL expression is a promising strategy in gene therapy for treatment of cancer.
Collapse
Affiliation(s)
- Ying Zhang
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Tsinghua University, Beijing 100005, China
| | | | | | | | | | | |
Collapse
|
25
|
Van Vliet KM, Blouin V, Brument N, Agbandje-McKenna M, Snyder RO. The role of the adeno-associated virus capsid in gene transfer. Methods Mol Biol 2008; 437:51-91. [PMID: 18369962 PMCID: PMC7120696 DOI: 10.1007/978-1-59745-210-6_2] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Adeno-associated virus (AAV) is one of the most promising viral gene transfer vectors that has been shown to effect long-term gene expression and disease correction with low toxicity in animal models, and is well tolerated in human clinical trials. The surface of the AAV capsid is an essential component that is involved in cell binding, internalization, and trafficking within the targeted cell. Prior to developing a gene therapy strategy that utilizes AAV, the serotype should be carefully considered since each capsid exhibits a unique tissue tropism and transduction efficiency. Several approaches have been undertaken in an effort to target AAV vectors to specific cell types, including utilizing natural serotypes that target a desired cellular receptor, producing pseudotyped vectors, and engineering chimeric and mosaic AAV capsids. These capsid modifications are being incorporated into vector production and purification methods that provide for the ability to scale-up the manufacturing process to support human clinical trials. Protocols for small-scale and large-scale production of AAV, as well as assays to characterize the final vector product, are presented here. The structures of AAV2, AAV4, and AAV5 have been solved by X-ray crystallography or cryo-electron microscopy (cryo-EM), and provide a basis for rational vector design in developing customized capsids for specific targeting of AAV vectors. The capsid of AAV has been shown to be remarkably stable, which is a desirable characteristic for a gene therapy vector; however, recently it has been shown that the AAV serotypes exhibit differential susceptibility to proteases. The capsid fragmentation pattern when exposed to various proteases, as well as the susceptibility of the serotypes to a series of proteases, provides a unique fingerprint for each serotype that can be used for capsid identity validation. In addition to serotype identification, protease susceptibility can also be utilized to study dynamic structural changes that must occur for the AAV capsid to perform its various functions during the virus life cycle. The use of proteases for structural studies in solution complements the crystal structural studies of the virus. A generic protocol based on proteolysis for AAV serotype identification is provided here.
Collapse
Affiliation(s)
- Kim M Van Vliet
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, USA
| | | | | | | | | |
Collapse
|
26
|
Wohlfahrt ME, Beard BC, Lieber A, Kiem HP. A capsid-modified, conditionally replicating oncolytic adenovirus vector expressing TRAIL Leads to enhanced cancer cell killing in human glioblastoma models. Cancer Res 2007; 67:8783-90. [PMID: 17875719 DOI: 10.1158/0008-5472.can-07-0357] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive brain tumor, and patients rarely survive for more than 2 years. Gene therapy may offer new treatment options and improve the prognosis for patients with GBM. Adenovirus-mediated gene therapy strategies for brain tumors have been limited by inefficient gene transfer due to low expression of the adenovirus serotype 5 (Ad5) receptor. We have used an adenovirus vector that specifically replicates in tumor cells and uses an Ad5 capsid and the adenovirus serotype (Ad35) fiber for efficient infection of malignant tumor cells. This vector also expresses adenovirus E1A and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in a tumor-specific manner. Here, we show that this oncolytic vector (Ad5/Ad35.IR-E1A/TRAIL) efficiently infects the GBM tumor cell lines SF767, T98G, and U-87 MG. Tumor cell killing was markedly enhanced with Ad5/Ad35.IR-E1A/TRAIL compared with wild-type Ad5 and Ad35 virus or Ad5/Ad35.IR-E1A- vectors without TRAIL expression in vitro. In vivo experiments using s.c. xenografted U-87 MG cells in NOD/SCID mice showed a significant growth delay of tumors after i.t. injection of Ad5/Ad35.IR-E1A/TRAIL, whereas adenovirus wild-type injections showed only marginal or no effect. Our findings indicate that the use of a capsid-modified adenoviral vector, in combination with TRAIL expression, is a promising novel approach for gene therapy of glioblastoma.
Collapse
Affiliation(s)
- Martin E Wohlfahrt
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109-1024, USA
| | | | | | | |
Collapse
|
27
|
Lin C, Wei W, Zhang J, Liu S, Liu Y, Zheng D. Formyl peptide receptor-like 1 mediated endogenous TRAIL gene expression with tumoricidal activity. Mol Cancer Ther 2007; 6:2618-25. [DOI: 10.1158/1535-7163.mct-07-0286] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
28
|
Abstract
The application of gene transfer technologies to the treatment of cancer has led to the development of new experimental approaches like gene directed enzyme/pro-drug therapy (GDEPT), inhibition of oncogenes and restoration of tumor-suppressor genes. In addition, gene therapy has a big impact on other fields like cancer immunotherapy, anti-angiogenic therapy and virotherapy. These strategies are being evaluated for the treatment of primary and metastatic liver cancer and some of them have reached clinical phases. We present a review on the basis and the actual status of gene therapy approaches applied to liver cancer.
Collapse
|
29
|
Hwang LH. Gene therapy strategies for hepatocellular carcinoma. J Biomed Sci 2006; 13:453-68. [PMID: 16633742 DOI: 10.1007/s11373-006-9085-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2006] [Accepted: 03/23/2006] [Indexed: 01/25/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most frequent cancers worldwide. Effective therapy to this cancer is currently lacking, creating an urgent need for new therapeutic strategies for HCC. Gene therapy approach that relies on the transduction of cells with genetic materials, such as apoptotic genes, suicide genes, genes coding for antiangiogenic factors or immunomodulatory molecules, small interfering RNA (siRNA), or oncolytic viral vectors, may provide a promising strategy. The aforementioned strategies have been largely evaluated in the animal models with HCC or liver metastasis. Due to the diversity of vectors and therapeutic genes, being used alone or in combination, gene therapy approach may generate great beneficial effects to control the growth of tumors within the liver.
Collapse
Affiliation(s)
- Lih-Hwa Hwang
- Hepatitis Research Center, National Taiwan University Hospital and Graduate Institute of Microbiology, National Taiwan University College of Medicine, 7 Chung-Shan S. Road, Taipei 10016, Taiwan, R.O.C.
| |
Collapse
|
30
|
Warrington KH, Herzog RW. Treatment of human disease by adeno-associated viral gene transfer. Hum Genet 2006; 119:571-603. [PMID: 16612615 DOI: 10.1007/s00439-006-0165-6] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2006] [Accepted: 02/28/2006] [Indexed: 11/24/2022]
Abstract
During the past decade, in vivo administration of viral gene transfer vectors for treatment of numerous human diseases has been brought from bench to bedside in the form of clinical trials, mostly aimed at establishing the safety of the protocol. In preclinical studies in animal models of human disease, adeno-associated viral (AAV) vectors have emerged as a favored gene transfer system for this approach. These vectors are derived from a replication-deficient, non-pathogenic parvovirus with a single-stranded DNA genome. Efficient gene transfer to numerous target cells and tissues has been described. AAV is particularly efficient in transduction of non-dividing cells, and the vector genome persists predominantly in episomal forms. Substantial correction, and in some instances complete cure, of genetic disease has been obtained in animal models of hemophilia, lysosomal storage disorders, retinal diseases, disorders of the central nervous system, and other diseases. Therapeutic expression often lasted for months to years. Treatments of genetic disorders, cancer, and other acquired diseases are summarized in this review. Vector development, results in animals, early clinical experience, as well as potential hurdles and challenges are discussed.
Collapse
Affiliation(s)
- Kenneth H Warrington
- Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL 32615-9586, USA
| | | |
Collapse
|