1
|
Pham Q, Glicksman J, Chatterjee A. Chemical approaches to probe and engineer AAV vectors. NANOSCALE 2024; 16:13820-13833. [PMID: 38978480 PMCID: PMC11271820 DOI: 10.1039/d4nr01300j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 06/14/2024] [Indexed: 07/10/2024]
Abstract
Adeno-associated virus (AAV) has emerged as the most promising vector for in vivo human gene therapy, with several therapeutic approvals in the last few years and countless more under development. Underlying this remarkable success are several attractive features that AAV offers, including lack of pathogenicity, low immunogenicity, long-term gene expression without genomic integration, the ability to infect both dividing and non-dividing cells, etc. However, the commonly used wild-type AAV capsids in therapeutic development present significant challenges, including inadequate tissue specificity and the need for large doses to attain therapeutic effectiveness, raising safety concerns. Additionally, significant preexisting adaptive immunity against most natural capsids, and the development of such anti-capsid immunity after the first treatment, represent major challenges. Strategies to engineer the AAV capsid are critically needed to address these challenges and unlock the full promise of AAV gene therapy. Chemical modification of the AAV capsid has recently emerged as a powerful new approach to engineer its properties. Unlike genetic strategies, which can be more disruptive to the delicate capsid assembly and packaging processes, "late-stage" chemical modification of the assembled capsid-whether at natural amino acid residues or site-specifically installed noncanonical amino acid residues-often enables a versatile approach to introducing new properties to the capsid. This review summarizes the significant recent progress in AAV capsid engineering strategies, with a particular focus on chemical modifications in advancing the next generation of AAV-based gene therapies.
Collapse
Affiliation(s)
- Quan Pham
- Department of Chemistry, Boston College, 2609 Beacon Street, Chestnut Hill, MA 02467, USA.
| | - Jake Glicksman
- Department of Chemistry, Boston College, 2609 Beacon Street, Chestnut Hill, MA 02467, USA.
| | - Abhishek Chatterjee
- Department of Chemistry, Boston College, 2609 Beacon Street, Chestnut Hill, MA 02467, USA.
| |
Collapse
|
2
|
Xiong J, Ling J, Yan J, Duan Y, Yu J, Li W, Yu W, Gao J, Xie D, Liu Z, Deng Y, Liao Y. LILRB4 knockdown inhibits aortic dissection development by regulating pyroptosis and the JAK2/STAT3 signaling pathway. Sci Rep 2024; 14:15564. [PMID: 38971897 PMCID: PMC11227527 DOI: 10.1038/s41598-024-66482-3] [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: 01/29/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024] Open
Abstract
Aortic dissection (AD) is a life-threatening condition with a high mortality rate and without effective pharmacological therapies. Our previous study illustrated that leukocyte immunoglobulin-like receptor B4 (LILRB4) knockdown promoted the contractile phenotypic switch and apoptosis of AD cells. This study aimed to further investigate the role of LILRB4 in animal models of AD and elucidate its underlying molecular mechanisms. Animal models of AD were established using 0.1% beta-aminopropionitrile and angiotensin II and an in vitro model was developed using platelet-derived growth factor BB (PDGF-BB). The effects of LILRB4 knockdown on histopathological changes, pyroptosis, phenotype transition, extracellular matrix (ECM), and Janus kinase 2 (JAK2)/signal transducers and activators of transcription 3 (STAT3) pathways were assessed using a series of in vivo and in vitro assays. The effects of the JAK2 inhibitor AG490 on AD cell function, phenotypic transition, and ECM were explored. LILRB4 was highly expressed in AD and its knockdown increased survival rate, reduced AD incidence, and alleviated histopathological changes in the AD mouse model. Furthermore, LILRB4 knockdown promoted contractile phenotype switch, stabilized the ECM, and inhibited pyroptosis. Mechanistically, LILRB4 knockdown inhibited the JAK2/STAT3 signaling pathway. JAK2 inhibitor AG490 inhibited cell viability and migration, enhanced apoptosis, induced G0/G1 cell cycle arrest, and suppressed S-phase progression in PDGF-BB-stimulated human aortic smooth muscle cells. LILRB4 knockdown suppresses AD development by inhibiting pyroptosis and the JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Jianxian Xiong
- Department of Cardiovascular Surgery, First Affiliated Hospital of Gannan Medical University, No. 23, Qingnian Road, Zhanggong District, Ganzhou City, 341000, Jiangxi Province, China
- Heart Medical Centre, First Affiliated Hospital of Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, China
| | - Jiayuan Ling
- Department of Cardiology, First Affiliated Hospital of Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, China
| | - Jie Yan
- Department of Thoracic Surgery, Nankang District First People's Hospital, Ganzhou City, 341400, Jiangxi Province, China
| | - Yanyu Duan
- Heart Medical Centre, First Affiliated Hospital of Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, China
- Engineering Research Center of Intelligent Acoustic Signals of Jiangxi Province, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, China
- Ganzhou Cardiovascular Rare Disease Diagnosis and Treatment Technology Innovation Center, Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, China
| | - Junjian Yu
- Department of Cardiovascular Surgery, First Affiliated Hospital of Gannan Medical University, No. 23, Qingnian Road, Zhanggong District, Ganzhou City, 341000, Jiangxi Province, China
- Heart Medical Centre, First Affiliated Hospital of Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, China
| | - Wentong Li
- Department of Cardiovascular Surgery, First Affiliated Hospital of Gannan Medical University, No. 23, Qingnian Road, Zhanggong District, Ganzhou City, 341000, Jiangxi Province, China
- Heart Medical Centre, First Affiliated Hospital of Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, China
| | - Wenbo Yu
- The First Clinical Medical College, Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, China
| | - Jianfeng Gao
- The First Clinical Medical College, Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, China
| | - Dilin Xie
- The First Clinical Medical College, Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, China
| | - Ziyou Liu
- Department of Cardiovascular Surgery, First Affiliated Hospital of Gannan Medical University, No. 23, Qingnian Road, Zhanggong District, Ganzhou City, 341000, Jiangxi Province, China.
- Heart Medical Centre, First Affiliated Hospital of Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, China.
| | - Yongzhi Deng
- Department of Cardiovascular Surgery, The Affiliated Hospital of Shanxi Medical University, Shanxi Cardiovascular Hospital (Institute), Shanxi Clinical Medical Research Center for Cardiovascular Disease, Taiyuan, 030024, China.
| | - Yongling Liao
- Heart Medical Centre, First Affiliated Hospital of Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, China.
- Department of Cardiology, First Affiliated Hospital of Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, China.
| |
Collapse
|
3
|
Macdonald J, Marx J, Büning H. Capsid-Engineering for Central Nervous System-Directed Gene Therapy with Adeno-Associated Virus Vectors. Hum Gene Ther 2021; 32:1096-1119. [PMID: 34662226 DOI: 10.1089/hum.2021.169] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Closing the gap in knowledge on the cause of neurodegenerative disorders is paving the way toward innovative treatment strategies, among which gene therapy has emerged as a top candidate. Both conventional gene therapy and genome editing approaches are being developed, and a great number of human clinical trials are ongoing. Already 2 years ago, the first gene therapy for a neurodegenerative disease, spinal muscular atrophy type 1 (SMA1), obtained market approval. To realize such innovative strategies, gene therapy delivery tools are key assets. Here, we focus on recombinant adeno-associated virus (AAV) vectors and report on strategies to improve first-generation vectors. Current efforts focus on the viral capsid to modify the host-vector interaction aiming at increasing the efficacy of target cell transduction, at simplifying vector administration, and at reducing the risk of vector dose-related side effects.
Collapse
Affiliation(s)
- Josephine Macdonald
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Jennifer Marx
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Hildegard Büning
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| |
Collapse
|
4
|
Jackson CB, Richard AS, Ojha A, Conkright KA, Trimarchi JM, Bailey CC, Alpert MD, Kay MA, Farzan M, Choe H. AAV vectors engineered to target insulin receptor greatly enhance intramuscular gene delivery. Mol Ther Methods Clin Dev 2020; 19:496-506. [PMID: 33313337 PMCID: PMC7710509 DOI: 10.1016/j.omtm.2020.11.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 11/11/2020] [Indexed: 12/22/2022]
Abstract
Adeno-associated virus (AAV) is one of the most commonly used vectors for gene therapy, and the applications for AAV-delivered therapies are numerous. However, the current state of technology is limited by the low efficiency with which most AAV vectors transduce skeletal muscle tissue. We demonstrate that vector efficiency can be enhanced by modifying the AAV capsid with a peptide that binds a receptor highly expressed in muscle tissue. When an insulin-mimetic peptide, S519, previously characterized for its high affinity to insulin receptor (IR), was inserted into the capsid, the AAV9 transduction efficiency of IR-expressing cell lines as well as differentiated primary human muscle cells was dramatically enhanced. This vector also exhibited efficient transduction of mouse muscle in vivo, resulting in up to 18-fold enhancement over AAV9. Owing to its superior transduction efficiency in skeletal muscle, we named this vector "enhanced AAV9" (eAAV9). We also found that the modification enhanced the transduction efficiency of several other AAV serotypes. Together, these data show that AAV transduction of skeletal muscle can be improved by targeting IR. They also show the broad utility of this modular strategy and suggest that it could also be applied to next-generation vectors that have yet to be engineered.
Collapse
Affiliation(s)
- Cody B. Jackson
- Department of Immunology and Microbiology, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Audrey S. Richard
- Department of Immunology and Microbiology, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Amrita Ojha
- Department of Immunology and Microbiology, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | | | | | | | | | - Mark A. Kay
- Departments of Pediatrics and Genetics, Stanford University, 269 Campus Dr. Rm 2105, Stanford, CA 94305, USA
| | - Michael Farzan
- Department of Immunology and Microbiology, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Hyeryun Choe
- Department of Immunology and Microbiology, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| |
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
|
Álvarez JV, Bravo SB, García-Vence M, De Castro MJ, Luzardo A, Colón C, Tomatsu S, Otero-Espinar FJ, Couce ML. Proteomic Analysis in Morquio A Cells Treated with Immobilized Enzymatic Replacement Therapy on Nanostructured Lipid Systems. Int J Mol Sci 2019; 20:ijms20184610. [PMID: 31540344 PMCID: PMC6769449 DOI: 10.3390/ijms20184610] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 09/07/2019] [Accepted: 09/13/2019] [Indexed: 12/14/2022] Open
Abstract
Morquio A syndrome, or mucopolysaccharidosis type IVA (MPS IVA), is a lysosomal storage disease due to mutations in the N-acetylgalactosamine-6-sulfatase (GALNS) gene. Systemic skeletal dysplasia and the related clinical features of MPS IVA are due to disruption of cartilage and its extracellular matrix, leading to an imbalance of growth. Enzyme replacement therapy (ERT) with recombinant human GALNS, alpha elosulfase, provides a systemic treatment. However, this therapy has a limited impact on skeletal dysplasia because the infused enzyme cannot penetrate cartilage and bone. Therefore, an alternative therapeutic approach to reach the cartilage is an unmet challenge. We have developed a new drug delivery system based on a nanostructure lipid carrier with the capacity to immobilize enzymes used for ERT and to target the lysosomes. This study aimed to assess the effect of the encapsulated enzyme in this new delivery system, using in vitro proteomic technology. We found a greater internalization of the enzyme carried by nanoparticles inside the cells and an improvement of cellular protein routes previously impaired by the disease, compared with conventional ERT. This is the first qualitative and quantitative proteomic assay that demonstrates the advantages of a new delivery system to improve the MPS IVA ERT.
Collapse
Affiliation(s)
- J Víctor Álvarez
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, Campus Vida, University of Santiago de Compostela, 15872 Santiago de Compostela, Spain.
- Department of Forensic Sciences, Pathology, Gynecology and Obstetrics, Pediatrics, Neonatology Service, Department of Paediatrics, Hospital Clínico Universitario de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), CIBERER, MetabERN, 15706 Santiago de Compostela, Spain.
- Skeletal Dysplasia Lab Nemours Biomedical Research Nemours/Alfred I. duPont Hospital for Children, 1600 Rockland Road, Wilmington, DE 19803, USA.
| | - Susana B Bravo
- Proteomic Platform, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario de Santiago de Compostela, 15706 Santiago de Compostea, Spain.
| | - María García-Vence
- Proteomic Platform, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario de Santiago de Compostela, 15706 Santiago de Compostea, Spain.
| | - María J De Castro
- Department of Forensic Sciences, Pathology, Gynecology and Obstetrics, Pediatrics, Neonatology Service, Department of Paediatrics, Hospital Clínico Universitario de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), CIBERER, MetabERN, 15706 Santiago de Compostela, Spain.
| | - Asteria Luzardo
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Sciences, Campus de Lugo, University of Santiago de Compostela, 27002 Lugo, Spain.
- Paraquasil Platform, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario de Santiago de Compostela, 15706 Santiago de Compostela, Spain.
| | - Cristóbal Colón
- Department of Forensic Sciences, Pathology, Gynecology and Obstetrics, Pediatrics, Neonatology Service, Department of Paediatrics, Hospital Clínico Universitario de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), CIBERER, MetabERN, 15706 Santiago de Compostela, Spain.
| | - Shunji Tomatsu
- Skeletal Dysplasia Lab Nemours Biomedical Research Nemours/Alfred I. duPont Hospital for Children, 1600 Rockland Road, Wilmington, DE 19803, USA.
| | - Francisco J Otero-Espinar
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, Campus Vida, University of Santiago de Compostela, 15872 Santiago de Compostela, Spain.
- Paraquasil Platform, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario de Santiago de Compostela, 15706 Santiago de Compostela, Spain.
| | - María L Couce
- Department of Forensic Sciences, Pathology, Gynecology and Obstetrics, Pediatrics, Neonatology Service, Department of Paediatrics, Hospital Clínico Universitario de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), CIBERER, MetabERN, 15706 Santiago de Compostela, Spain.
| |
Collapse
|
7
|
Eichhoff AM, Börner K, Albrecht B, Schäfer W, Baum N, Haag F, Körbelin J, Trepel M, Braren I, Grimm D, Adriouch S, Koch-Nolte F. Nanobody-Enhanced Targeting of AAV Gene Therapy Vectors. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 15:211-220. [PMID: 31687421 PMCID: PMC6819893 DOI: 10.1016/j.omtm.2019.09.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 09/10/2019] [Indexed: 12/12/2022]
Abstract
A limiting factor for the use of adeno-associated viruses (AAVs) as vectors in gene therapy is the broad tropism of AAV serotypes, i.e., the parallel infection of several cell types. Nanobodies are single immunoglobulin variable domains from heavy chain antibodies that naturally occur in camelids. Their small size and high solubility allow easy reformatting into fusion proteins. Herein we show that a membrane protein-specific nanobody can be inserted into a surface loop of the VP1 capsid protein of AAV2. Using three structurally distinct membrane proteins—a multispan ion channel, a single-span transmembrane protein, and a glycosylphosphatidylinositol (GPI)-anchored ectoenzyme—we show that this strategy can dramatically enhance the transduction of specific target cells by recombinant AAV2. Moreover, we show that the nanobody-VP1 fusion of AAV2 can be incorporated into the capsids of AAV1, AAV8, and AAV9 and thereby effectively redirect the target specificity of other AAV serotypes. Nanobody-mediated targeting provides a highly efficient AAV targeting strategy that is likely to open up new avenues for genetic engineering of cells.
Collapse
Affiliation(s)
- Anna Marei Eichhoff
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Institute for Research and Innovation in Biomedicine, INSERM U1234, Normandy University, Rouen, France
| | - Kathleen Börner
- Department of Infectious Diseases/Virology, Heidelberg University Hospital, Heidelberg, Germany.,BioQuant, Heidelberg University, Heidelberg, Germany.,German Center for Infection Research, Heidelberg, Germany
| | - Birte Albrecht
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Waldemar Schäfer
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Natalie Baum
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Haag
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jakob Körbelin
- Center for Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martin Trepel
- Center for Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ingke Braren
- Institute of Pharmacology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dirk Grimm
- Department of Infectious Diseases/Virology, Heidelberg University Hospital, Heidelberg, Germany.,BioQuant, Heidelberg University, Heidelberg, Germany.,German Center for Infection Research, Heidelberg, Germany
| | - Sahil Adriouch
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Institute for Research and Innovation in Biomedicine, INSERM U1234, Normandy University, Rouen, France
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
8
|
Cao J, Liu X, Yuan Y, Wang F, Kong W, Shi G, Li W, Zhang C. A rAAV2/6 Mutant with Enhanced Targeting for Mouse Retinal Müller Cells. Curr Eye Res 2019; 45:64-71. [PMID: 31294618 DOI: 10.1080/02713683.2019.1639768] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Purpose: Adeno-associated virus vector (AAV) is the most accepted gene delivery vector for retinal gene therapy. Müller cells play an important role in maintaining homeostasis and neuronal structural integrity, stability and it has been found to be involved in many retinopathies. The aim of this study is to identify a rAAV2/6 mutant which has increased tropism for Müller cell of the mouse retina.Materials and Methods: Using amino acid mutagenesis, we created a rAAV2/6 capsid mutant, rAAV2/6-S663L. In vivo imaging and retinal flat mount were employed to analyze the gene expression of rAAV2/6-S663L and wt rAAV2/6 in mouse retinal tissue. Retinal tissue cryosection, immunohistochemistry (IHC), Müller cell-specific promoter-controlled gene expression, and double AAV fluorescent protein co-expression were performed to determine the targeting of rAAV2/6-S663L for mouse retinal Müller cells.Results: In vivo imaging, retinal flat mount and retinal tissue cryosection results showed that rAAV2/6-S663L and wt rAAV2/6 have different specific tropisms in mouse retina and rAAV2/6-S663L is more preferentially targeting Müller cells. Müller cell-specific promoter-controlled gene expression experiments and IHC test confirmed that rAAV2/6-S663L has a higher tendency to infect Müller cells than wt rAAV2/6. Co-infection of the mouse retina with one rAAV2/6-S663L expressing EGFP under the control of GFAP promoter and the other one expressing mCherry under the control of CMV promoter revealed co-expression of the two fluorescent proteins in Müller cells.Conclusions: The results confirmed that rAAV2/6-S663L has a higher tropism for Müller cells than wt rAAV2/6. Our findings could add a new useful tool for retinal disease gene therapy.
Collapse
Affiliation(s)
- Jinjing Cao
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China.,Suzhou Institute of Biomedical Engineering and Technology, University of Science and Technology of China, Suzhou, China
| | - Xiaomei Liu
- Suzhou Institute of Biomedical Engineering and Technology, University of Science and Technology of China, Suzhou, China
| | - Yun Yuan
- Suzhou Institute of Biomedical Engineering and Technology, University of Science and Technology of China, Suzhou, China
| | - Feifei Wang
- Suzhou Institute of Biomedical Engineering and Technology, University of Science and Technology of China, Suzhou, China
| | - Wen Kong
- Suzhou Institute of Biomedical Engineering and Technology, University of Science and Technology of China, Suzhou, China
| | - Guohua Shi
- Suzhou Institute of Biomedical Engineering and Technology, University of Science and Technology of China, Suzhou, China.,The Innovation Center of Excellence on Brain Science, Chinese Academy of Sciences, Shanghai, China
| | - Wensheng Li
- Aier School of Ophthalmology, Central South University, Changsha, China.,Ophthalmology, Shanghai Aier Eye Hospital, Shanghai, China
| | - Chun Zhang
- Suzhou Institute of Biomedical Engineering and Technology, University of Science and Technology of China, Suzhou, China
| |
Collapse
|
9
|
Mietzsch M, Pénzes JJ, Agbandje-McKenna M. Twenty-Five Years of Structural Parvovirology. Viruses 2019; 11:E362. [PMID: 31010002 PMCID: PMC6521121 DOI: 10.3390/v11040362] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/10/2019] [Accepted: 04/11/2019] [Indexed: 12/13/2022] Open
Abstract
Parvoviruses, infecting vertebrates and invertebrates, are a family of single-stranded DNA viruses with small, non-enveloped capsids with T = 1 icosahedral symmetry. A quarter of a century after the first parvovirus capsid structure was published, approximately 100 additional structures have been analyzed. This first structure was that of Canine Parvovirus, and it initiated the practice of structure-to-function correlation for the family. Despite high diversity in the capsid viral protein (VP) sequence, the structural topologies of all parvoviral capsids are conserved. However, surface loops inserted between the core secondary structure elements vary in conformation that enables the assembly of unique capsid surface morphologies within individual genera. These variations enable each virus to establish host niches by allowing host receptor attachment, specific tissue tropism, and antigenic diversity. This review focuses on the diversity among the parvoviruses with respect to the transcriptional strategy of the encoded VPs, the advances in capsid structure-function annotation, and therapeutic developments facilitated by the available structures.
Collapse
Affiliation(s)
- Mario Mietzsch
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, The McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA.
| | - Judit J Pénzes
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, The McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA.
| | - Mavis Agbandje-McKenna
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, The McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
10
|
Finet JE, Wan X, Donahue JK. Fusion of Anthopleurin-B to AAV2 increases specificity of cardiac gene transfer. Virology 2018; 513:43-51. [PMID: 29032346 DOI: 10.1016/j.virol.2017.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 09/27/2017] [Accepted: 10/03/2017] [Indexed: 10/18/2022]
Abstract
AAV-mediated gene therapy has become a promising therapeutic strategy for chronic diseases. Its clinical utilization, however, is limited by the potential risk of off-target effects. In this work we attempt to overcome this challenge, hypothesizing that cardiac ion channel-specific ligands could be fused onto the AAV capsid, and narrow its tropism to cardiac myocytes. We successfully fused the cardiac sodium channel (Nav1.5)-binding toxin Anthopleurin-B onto the AAV2 capsid without compromising virus integrity, and demonstrated increased specificity of cardiomyocyte attachment. Although virus attachment to Nav1.5 did not supersede the natural heparan-mediated virus binding, heparan-binding ablated vectors carrying Anthopleurin-B eliminated hepatic and other extracardiac gene transfer, while preserving cardiac myocyte gene transfer. Virus binding to the cardiac sodium channel transiently decreased sodium current density, but did not cause any arrhythmias. Our findings expand the knowledge of attachment, infectivity, and intracellular processing of AAV vectors, and present an alternative strategy for vector retargeting.
Collapse
Affiliation(s)
- J Emanuel Finet
- Krannert Institute of Cardiology, Department of Medicine, Indiana University, Indianapolis, IN, USA
| | - Xiaoping Wan
- Heart and Vascular Research Center, Case Western Reserve University, MetroHealth Campus, Cleveland, OH, USA
| | - J Kevin Donahue
- Division of Cardiology, Department of Medicine, University of Massachusetts Medical School, 368 Plantation Street, Albert Sherman Center, 7th floor, Worcester, MA 01605, USA.
| |
Collapse
|
11
|
Wu Y, Zhang L, Cui C, Cansiz S, Liang H, Wu C, Teng IT, Chen W, Liu Y, Hou W, Zhang X, Tan W. Enhanced Targeted Gene Transduction: AAV2 Vectors Conjugated to Multiple Aptamers via Reducible Disulfide Linkages. J Am Chem Soc 2017; 140:2-5. [PMID: 29256602 DOI: 10.1021/jacs.7b08518] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Enhanced targeted gene transduction by AAV2 vectors is achieved by linking the vector to multiple sgc8 aptamers, which are selective for cell membrane protein PTK7. Aptamer molecules are conjugated to multiple sites on a DNA dendrimer (G-sgc8), which is then linked to AAV2 via a dithiobis(succinimidyl propionate) cross-linker containing a disulfide group, which can facilitate the release of AAV2 vectors by reaction with the reduced form of intracellular glutathione. The G-sgc8-AAV2 vectors showed a 21-fold enhancement in binding affinity and an enhanced ability to protect sgc8 aptamers against nuclease degradation to cells expressing PTK7 compared to single aptamer-AAV2 conjugates. The transduction efficiency was tested by loading AAV2 with the gene for green fluorescent protein. Therefore, this modified recombinant vector is an attractive and promising tool for targeted biomedical applications.
Collapse
Affiliation(s)
- Yuan Wu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Life Sciences, and Aptamer Engineering Center of Hunan Province, Hunan University , Changsha, Hunan 410082, China.,Center for Research at Bio/Nano Interface, Department of Chemistry and Department of Physiology and Functional Genomics, University Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida , Gainesville, Florida 32611-7200, United States
| | - Liqin Zhang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Life Sciences, and Aptamer Engineering Center of Hunan Province, Hunan University , Changsha, Hunan 410082, China.,Center for Research at Bio/Nano Interface, Department of Chemistry and Department of Physiology and Functional Genomics, University Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida , Gainesville, Florida 32611-7200, United States
| | - Cheng Cui
- Center for Research at Bio/Nano Interface, Department of Chemistry and Department of Physiology and Functional Genomics, University Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida , Gainesville, Florida 32611-7200, United States
| | - Sena Cansiz
- Center for Research at Bio/Nano Interface, Department of Chemistry and Department of Physiology and Functional Genomics, University Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida , Gainesville, Florida 32611-7200, United States
| | - Hao Liang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Life Sciences, and Aptamer Engineering Center of Hunan Province, Hunan University , Changsha, Hunan 410082, China
| | - Cuichen Wu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Life Sciences, and Aptamer Engineering Center of Hunan Province, Hunan University , Changsha, Hunan 410082, China.,Center for Research at Bio/Nano Interface, Department of Chemistry and Department of Physiology and Functional Genomics, University Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida , Gainesville, Florida 32611-7200, United States
| | - I-Ting Teng
- Center for Research at Bio/Nano Interface, Department of Chemistry and Department of Physiology and Functional Genomics, University Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida , Gainesville, Florida 32611-7200, United States
| | - Weijun Chen
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Life Sciences, and Aptamer Engineering Center of Hunan Province, Hunan University , Changsha, Hunan 410082, China.,Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University , Shanghai 200240, China.,Center for Research at Bio/Nano Interface, Department of Chemistry and Department of Physiology and Functional Genomics, University Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida , Gainesville, Florida 32611-7200, United States
| | - Yuan Liu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Life Sciences, and Aptamer Engineering Center of Hunan Province, Hunan University , Changsha, Hunan 410082, China.,Center for Research at Bio/Nano Interface, Department of Chemistry and Department of Physiology and Functional Genomics, University Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida , Gainesville, Florida 32611-7200, United States
| | - Weijia Hou
- Center for Research at Bio/Nano Interface, Department of Chemistry and Department of Physiology and Functional Genomics, University Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida , Gainesville, Florida 32611-7200, United States
| | - Xiaobing Zhang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Life Sciences, and Aptamer Engineering Center of Hunan Province, Hunan University , Changsha, Hunan 410082, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Life Sciences, and Aptamer Engineering Center of Hunan Province, Hunan University , Changsha, Hunan 410082, China.,Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University , Shanghai 200240, China.,Center for Research at Bio/Nano Interface, Department of Chemistry and Department of Physiology and Functional Genomics, University Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida , Gainesville, Florida 32611-7200, United States
| |
Collapse
|
12
|
Rey-Rico A, Cucchiarini M. Controlled release strategies for rAAV-mediated gene delivery. Acta Biomater 2016; 29:1-10. [PMID: 26472612 DOI: 10.1016/j.actbio.2015.10.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 10/02/2015] [Accepted: 10/09/2015] [Indexed: 12/17/2022]
Abstract
The development of efficient and safe gene transfer vectors capable of achieving appropriate levels of therapeutic gene expression in a target is one of the most challenging issues in clinical gene therapy. Diverse nonviral and viral gene vehicles have been developed to modify human cells and tissues that may be affected in a variety of diseases, among which the nonpathogenic, effective, and relatively safe recombinant adeno-associated viral (rAAV) vectors that make them a preferred gene delivery system to treat human disorders. Yet, their adapted clinical application is still limited by several hurdles including the presence of immune responses in the host organism and the existence of rate-limiting steps associated with physiological barriers. The use of controlled release strategies to deliver gene vectors such as rAAV may provide powerful tools to enhance the temporal and spatial presentation of therapeutic agents in a defined target and to overcome such obstacles in vivo. The goal of this review is to provide an overview of the most recent advances in gene therapy with a focus on rAAV vectors for clinical translation based on the controlled release from adapted biomaterials as a means to improve the performance of the gene transfer procedure. We also discuss the challenges that remain to be addressed for a safe and efficient adaptation and use of such approaches in the patient. STATEMENT OF SIGNIFICANCE The development of effective gene vectors to achieve suitable levels of a therapeutic agent in a target is a critical issue in clinical gene therapy and regenerative medicine. Diverse vehicles are currently available among which the nonpathogenic recombinant adeno-associated virus (rAAV) vectors, a preferred system to effectively treat human disorders. Yet, the clinical use of rAAV is impaired by the host immune responses and by rate-limiting steps of transgene expression. Controlled rAAV delivery systems may provide workable approaches to overcome such obstacles. Here, we give an overview of the most recent advances on the controlled release of vectors with a focus on rAAV using adapted biomaterials and discuss the key challenges for a safe translation in patients.
Collapse
|
13
|
Büning H, Huber A, Zhang L, Meumann N, Hacker U. Engineering the AAV capsid to optimize vector–host-interactions. Curr Opin Pharmacol 2015; 24:94-104. [DOI: 10.1016/j.coph.2015.08.002] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 08/03/2015] [Accepted: 08/05/2015] [Indexed: 12/15/2022]
|
14
|
Abstract
BACKGROUND Gene therapy for inherited retinal diseases (IRDs) is currently being validated in several clinical trials and is becoming a promising therapeutic option for these previously incurable diseases. OBJECTIVES The aim of this review is to give an overview of the concept, the application and the challenges associated with gene therapy. In particular, the pertinence of gene therapy for IRDs will be highlighted along with ongoing clinical trials in the field. MATERIAL AND METHODS A systematic review of relevant entries on gene therapy and on gene therapy for IRDs, in particular in PubMed and ClinicalTrials.gov. RESULTS Gene therapy is emerging not only as a therapy for monogenetic retinal diseases but also for complex genetic diseases, such as neovascular age-related macular degeneration. The discovery of adeno-associated viral vectors (AAVs) has marked a great improvement for IRD gene therapy. All clinical studies since 2006 demonstrated the safety and initial efficacy; however, not all expectations based on very successful preclinical studies were met. CONCLUSION In future we can expect gene therapy to continue to become more clinically relevant. More than ever, it is now essential to generate precise characterizations of the natural disease progression of IRDs through observational or retrospective studies in order to guarantee a most effective study design.
Collapse
Affiliation(s)
- J-S Bellingrath
- Universitäts-Augenklinik, Department für Augenheilkunde, Universitätsklinikum Tübingen, Schleichstr. 12-16, 72076, Tübingen, Deutschland
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Headley Way, Oxford, OX3 9DU,, England
| | - M D Fischer
- Universitäts-Augenklinik, Department für Augenheilkunde, Universitätsklinikum Tübingen, Schleichstr. 12-16, 72076, Tübingen, Deutschland.
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Headley Way, Oxford, OX3 9DU,, England.
| |
Collapse
|
15
|
Voráčková I, Ulbrich P, Diehl WE, Ruml T. Engineered retroviral virus-like particles for receptor targeting. Arch Virol 2013; 159:677-88. [DOI: 10.1007/s00705-013-1873-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 09/30/2013] [Indexed: 10/26/2022]
|
16
|
Tomatsu S, Alméciga-Díaz CJ, Barbosa H, Montaño AM, Barrera LA, Shimada T, Yasuda E, Mackenzie WG, Mason RW, Suzuki Y, Orii KE, Orii T. Therapies of mucopolysaccharidosis IVA (Morquio A syndrome). Expert Opin Orphan Drugs 2013; 1:805-818. [PMID: 25419501 DOI: 10.1517/21678707.2013.846853] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
INTRODUCTION Morquio A syndrome (mucopolysaccharidosis type IVA, MPS IVA) is one of the lysosomal storage diseases and is caused by the deficiency of N-acetylgalactosamine-6-sulfate sulfatase (GALNS). Deficiency of this enzyme leads to accumulation of glycosaminoglycans (GAGs), keratan sulfate (KS) and chondroitin-6-sulfate (C6S). The majority of KS is produced by chondrocytes, and therefore, the undegraded substrates accumulate mainly in cells and extracelluar matrix (ECM) of cartilage. This has a direct impact on cartilage and bone development, leading to systemic skeletal dysplasia. In patients with Morquio A, cartilage cells are vacuolated, and this results in abnormal chondrogenesis and/or endochondral ossification. AREAS COVERED This article describes the advanced therapies of Morquio A, focused on enzyme replacement therapy (ERT) and gene therapy to deliver the drug to avascular bone lesions. ERT and gene therapies for other types of MPS are also discussed, which provide therapeutic efficacy to bone lesions. EXPERT OPINION ERT, gene therapy and hematopietic stem therapy are clinically and/or experimentally conducted. However, there is no effective curative therapy for bone lesion to date. One of the limitations for Morquio A therapy is that targeting avascular cartilage tissues remains an unmet challenge. ERT or gene therapy with bone-targeting system will improve the bone pathology and skeletal manifestations more efficiently.
Collapse
Affiliation(s)
- Shunji Tomatsu
- Professor and Director, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA ; Nemours/Alfred I. duPont Hospital for Children, Skeletal Dysplasia Center, Nemours Biomedical Research, 1600 Rockland Rd., Wilmington, DE 19803, USA
| | - Carlos J Alméciga-Díaz
- Institute for the Study of Inborn Errors of Metabolism, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - Hector Barbosa
- Institute for the Study of Inborn Errors of Metabolism, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - Adriana M Montaño
- Saint Louis University, Department of Pediatrics, St. Louis, MO, USA
| | - Luis A Barrera
- Institute for the Study of Inborn Errors of Metabolism, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - Tsutomu Shimada
- Professor and Director, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Eriko Yasuda
- Professor and Director, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - William G Mackenzie
- Professor and Director, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Robert W Mason
- Professor and Director, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Yasuyuki Suzuki
- Gifu University, Medical Education Development Center, Gifu, Japan
| | - Kenji E Orii
- Gifu University, Department of Pediatrics, Gifu, Japan
| | - Tadao Orii
- Gifu University, Department of Pediatrics, Gifu, Japan
| |
Collapse
|
17
|
Chen C, Akerstrom V, Baus J, Lan MS, Breslin MB. Comparative analysis of the transduction efficiency of five adeno associated virus serotypes and VSV-G pseudotype lentiviral vector in lung cancer cells. Virol J 2013; 10:86. [PMID: 23497017 PMCID: PMC3602085 DOI: 10.1186/1743-422x-10-86] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 03/07/2013] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Lung cancer is the leading cause of cancer-related deaths in the US. Recombinant vectors based on adeno-associated virus (AAV) and lentivirus are promising delivery tools for gene therapy due to low toxicity and long term expression. The efficiency of the gene delivery system is one of the most important factors directly related to the success of gene therapy. METHODS We infected SCLC cell lines, SHP-77, DMS 53, NCI-H82, NCI-H69, NCI-H727, NCI-H1155, and NSCLC cell lines, NCI-H23, NCI-H661, and NCI-H460 with VSV-G pseudo-typed lentivirus or 5 AAV serotypes, AAV2/1, AAV2/2, AAV2/4, AAV2/5, and AAV2/8 expressing the CMV promoter mCherry or green fluorescent protein transgene (EGFP). The transduction efficiency was analyzed by fluorescent microscopy and flow cytometry. RESULTS Of all the serotypes of AAV examined, AAV2/1 was the optimal serotype in most of the lung cancer cell lines except for NCI-H69 and NCI-H82. The highest transduction rate achieved with AAV2/1 was between 30-50% at MOI 100. Compared to all AAV serotypes, lentivirus had the highest transduction efficiency of over 50% at MOI 1. Even in NCI-H69 cells resistant to all AAV serotypes, lentivirus had a 10-40% transduction rate. To date, AAV2 is the most widely-used serotype to deliver a transgene. Our results showed the transduction efficiency of AAVs tested was AAV2/1 > AA2/5 = AAV2/2> > AAV2/4 and AAV2/8. CONCLUSIONS This study demonstrated that VSV-G pseudotyped lentivirus and AAV2/1 can mediate expression of a transgene for lung cancer gene therapy. Overall, our results showed that lentivirus is the best candidate to deliver a transgene into lung cancer cells for treatment.
Collapse
Affiliation(s)
- Chiachen Chen
- Research Institute For Children, Children's Hospital New Orleans, New Orleans, LA 70118, USA
| | | | | | | | | |
Collapse
|
18
|
|
19
|
Lerch TF, O’Donnell JK, Meyer NL, Xie Q, Taylor KA, Stagg SM, Chapman MS. Structure of AAV-DJ, a retargeted gene therapy vector: cryo-electron microscopy at 4.5 Å resolution. Structure 2012; 20:1310-20. [PMID: 22727812 PMCID: PMC3418430 DOI: 10.1016/j.str.2012.05.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 05/10/2012] [Accepted: 05/11/2012] [Indexed: 12/19/2022]
Abstract
AAV-DJ, a leading candidate vector for liver gene therapy, was created through random homologous recombination followed by directed evolution, selecting for in vivo liver tropism and resistance to in vitro immune neutralization. Here, the 4.5 Å resolution cryo-EM structure is determined for the engineered AAV vector, revealing structural features that illuminate its phenotype. The heparan sulfate receptor-binding site is little changed from AAV-2, and heparin-binding affinity is similar. A loop that is antigenic in other serotypes has a unique conformation in AAV-DJ that would conflict with the binding of an AAV-2 neutralizing monoclonal antibody. This is consistent with increased resistance to neutralization by human polyclonal sera, raising the possibility that changed tropism may be a secondary effect of altered immune interactions. The reconstruction exemplifies analysis of fine structural changes and the potential of cryo-EM, in favorable cases, to characterize mutant or ligand-bound complexes.
Collapse
Affiliation(s)
- Thomas F. Lerch
- Department of Biochemistry & Molecular Biology, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jason K. O’Donnell
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
| | - Nancy L. Meyer
- Department of Biochemistry & Molecular Biology, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Qing Xie
- Department of Biochemistry & Molecular Biology, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Kenneth A. Taylor
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Scott M. Stagg
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
- Department of Chemistry & Biochemistry, Florida State University, Tallahassee, FL 32306, USA
| | - Michael S. Chapman
- Department of Biochemistry & Molecular Biology, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
20
|
Lerch TF, Chapman MS. Identification of the heparin binding site on adeno-associated virus serotype 3B (AAV-3B). Virology 2011; 423:6-13. [PMID: 22169623 DOI: 10.1016/j.virol.2011.10.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 09/27/2011] [Accepted: 10/10/2011] [Indexed: 12/12/2022]
Abstract
Adeno-associated virus is a promising vector for gene therapy. In the current study, the binding site on AAV serotype 3B for the heparan sulfate proteoglycan (HSPG) receptor has been characterized. X-ray diffraction identified a disaccharide binding site at the most positively charged region on the virus surface. The contributions of basic amino acids at this and other sites were characterized using site-directed mutagenesis. Both heparin and cell binding are correlated to positive charge at the disaccharide binding site, and transduction is significantly decreased in AAV-3B vectors mutated at this site to reduce heparin binding. While the receptor attachment sites of AAV-3B and AAV-2 are both in the general vicinity of the viral spikes, the exact amino acids that participate in electrostatic interactions are distinct. Diversity in the mechanisms of cell attachment by AAV serotypes will be an important consideration for the rational design of improved gene therapy vectors.
Collapse
Affiliation(s)
- Thomas F Lerch
- Department of Biochemistry & Molecular Biology, School of Medicine, Oregon Health & Science University, Portland, OR 97239-3098, USA
| | | |
Collapse
|
21
|
Kleinlogel S, Terpitz U, Legrum B, Gökbuget D, Boyden ES, Bamann C, Wood PG, Bamberg E. A gene-fusion strategy for stoichiometric and co-localized expression of light-gated membrane proteins. Nat Methods 2011; 8:1083-8. [DOI: 10.1038/nmeth.1766] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Accepted: 09/19/2011] [Indexed: 01/24/2023]
|
22
|
Cao H, Zhang GR, Geller AI. Antibody-mediated targeted gene transfer of helper virus-free HSV-1 vectors to rat neocortical neurons that contain either NMDA receptor 2B or 2A subunits. Brain Res 2011; 1415:127-35. [PMID: 21885042 PMCID: PMC3176983 DOI: 10.1016/j.brainres.2011.08.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Revised: 07/13/2011] [Accepted: 08/05/2011] [Indexed: 02/04/2023]
Abstract
Because of the numerous types of neurons in the brain, and particularly the forebrain, neuron type-specific expression will benefit many potential applications of direct gene transfer. The two most promising approaches for achieving neuron type-specific expression are targeted gene transfer to a specific type of neuron and using a neuron type-specific promoter. We previously developed antibody-mediated targeted gene transfer with Herpes Simplex Virus (HSV-1) vectors by modifying glycoprotein C (gC) to replace the heparin binding domain, which mediates the initial binding of HSV-1 particles to many cell types, with the Staphylococcus A protein ZZ domain, which binds immunoglobulin (Ig) G. We showed that a chimeric gC-ZZ protein is incorporated into vector particles and binds IgG. As a proof-of-principle for antibody-mediated targeted gene transfer, we isolated complexes of these vector particles and an anti-NMDA NR1 subunit antibody, and demonstrated targeted gene transfer to neocortical cells that contain NR1 subunits. However, because most forebrain neurons contain NR1, we obtained only a modest increase in the specificity of gene transfer, and this targeting specificity is of limited utility for physiological experiments. Here, we report efficient antibody-mediated targeted gene transfer to NMDA NR2B- or NR2A-containing cells in rat postrhinal cortex, and a neuron-specific promoter further restricted recombinant expression to neurons. Of note, because NR2A-containing neurons are relatively rare, these results show that antibody-mediated targeted gene transfer with HSV-1 vectors containing neuron type-specific promoters can restrict recombinant expression to specific types of forebrain neurons of physiological significance.
Collapse
Affiliation(s)
- Haiyan Cao
- Department of Neurology, West Roxbury VA Hospital/Harvard Medical School, W. Roxbury, MA 02132, USA
| | | | | |
Collapse
|
23
|
Martini SV, Rocco PRM, Morales MM. Adeno-associated virus for cystic fibrosis gene therapy. Braz J Med Biol Res 2011; 44:1097-104. [PMID: 21952739 DOI: 10.1590/s0100-879x2011007500123] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Accepted: 09/13/2011] [Indexed: 12/12/2022] Open
Abstract
Gene therapy is an alternative treatment for genetic lung disease, especially monogenic disorders such as cystic fibrosis. Cystic fibrosis is a severe autosomal recessive disease affecting one in 2500 live births in the white population, caused by mutation of the cystic fibrosis transmembrane conductance regulator (CFTR). The disease is classically characterized by pancreatic enzyme insufficiency, an increased concentration of chloride in sweat, and varying severity of chronic obstructive lung disease. Currently, the greatest challenge for gene therapy is finding an ideal vector to deliver the transgene (CFTR) to the affected organ (lung). Adeno-associated virus is the most promising viral vector system for the treatment of respiratory disease because it has natural tropism for airway epithelial cells and does not cause any human disease. This review focuses on the basic properties of adeno-associated virus and its use as a vector for cystic fibrosis gene therapy.
Collapse
Affiliation(s)
- S V Martini
- Laboratório de Fisiologia Celular e Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | |
Collapse
|
24
|
Gwathmey JK, Yerevanian AI, Hajjar RJ. Cardiac gene therapy with SERCA2a: from bench to bedside. J Mol Cell Cardiol 2011; 50:803-12. [PMID: 21093451 PMCID: PMC3075330 DOI: 10.1016/j.yjmcc.2010.11.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2010] [Revised: 10/27/2010] [Accepted: 11/10/2010] [Indexed: 10/18/2022]
Abstract
While progress in conventional treatments is making steady and incremental gains to reduce mortality associated with heart failure, there remains a need to explore potentially new therapeutic approaches. Heart failure induced by different etiologies such as coronary artery disease, hypertension, diabetes, infection, or inflammation results generally in calcium cycling dysregulation at the myocyte level. Recent advances in understanding of the molecular basis of these calcium cycling abnormalities, together with the evolution of increasingly efficient gene transfer technology, have placed heart failure within reach of gene-based therapy. Furthermore, the recent successful completion of a phase 2 trial targeting the sarcoplasmic reticulum calcium pump (SERCA2a) ushers in a new era for gene therapy for the treatment of heart failure. This article is part of a Special Section entitled "Special Section: Cardiovascular Gene Therapy".
Collapse
|
25
|
Abstract
The success of any gene transfer procedure, either through in vivo inoculation of the genetic material or after gene transfer into the patient’s cells ex vivo, strictly depends upon the efficiency of nucleic acid internalization by the target cells. As a matter of fact, making gene transfer more efficient continues to represent the most relevant challenge to the clinical success of gene therapy.
Collapse
Affiliation(s)
- Mauro Giacca
- grid.425196.d0000000417594810International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| |
Collapse
|
26
|
Wang J, Faust SM, Rabinowitz JE. The next step in gene delivery: molecular engineering of adeno-associated virus serotypes. J Mol Cell Cardiol 2010; 50:793-802. [PMID: 21029739 DOI: 10.1016/j.yjmcc.2010.10.017] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Revised: 10/12/2010] [Accepted: 10/13/2010] [Indexed: 12/29/2022]
Abstract
Delivery is at the heart of gene therapy. Viral DNA delivery systems are asked to avoid the immune system, transduce specific target cell types while avoiding other cell types, infect dividing and non-dividing cells, insert their cargo within the host genome without mutagenesis or to remain episomal, and efficiently express transgenes for a substantial portion of a lifespan. These sought-after features cannot be associated with a single delivery system, or can they? The Adeno-associated virus family of gene delivery vehicles has proven to be highly malleable. Pseudotyping, using AAV serotype 2 terminal repeats to generate designer shells capable of transducing selected cell types, enables the packaging of common genomes into multiple serotypes virions to directly compare gene expression and tropism. In this review the ability to manipulate this virus will be examined from the inside out. The influence of host cell factors and organism biology including the immune response on the molecular fate of the viral genome will be discussed as well as differences in cellular trafficking patterns and uncoating properties that influence serotype transduction. Re-engineering the prototype vector AAV2 using epitope insertion, chemical modification, and molecular evolution not only demonstrated the flexibility of the best-studied serotype, but now also expanded the tool kit for molecular modification of all AAV serotypes. Current AAV research has changed its focus from examination of wild-type AAV biology to the feedback of host cell/organism on the design and development of a new generation of recombinant AAV delivery vehicles. This article is part of a Special Section entitled "Special Section: Cardiovascular Gene Therapy".
Collapse
Affiliation(s)
- Jinhui Wang
- Thomas Jefferson University Center for Translational Medicine, Philadelphia, PA 19107, USA
| | | | | |
Collapse
|
27
|
Xie W, Liu S, Su H, Wang Z, Zheng Y, Fu Y. Ultrasound microbubbles enhance recombinant adeno-associated virus vector delivery to retinal ganglion cells in vivo. Acad Radiol 2010; 17:1242-8. [PMID: 20619698 DOI: 10.1016/j.acra.2010.05.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2010] [Revised: 05/08/2010] [Accepted: 05/10/2010] [Indexed: 01/10/2023]
Abstract
RATIONALE AND OBJECTIVES The aim of this study was to investigate whether ultrasound-mediated microbubble destruction enhances the transduction efficiency of recombinant adeno-associated virus (rAAV)-mediated enhanced green fluorescent protein (EGFP) gene into retinal ganglion cells (RGCs) of rats and whether it causes relevant adverse effects. MATERIALS AND METHODS Thirty-two adult Sprague-Dawley rats were divided into four groups with different ultrasound powers, and retinal flat mounts and hematoxylin and eosin staining sections were made for optimization of parameters. A further 70 adult Sprague-Dawley rats were divided into four groups randomly. The first group (group A) was used as a normal control with 10 rats, and the remaining rats were evenly divided into groups B, C, and D. Each group included 20 rats. Groups B and C received rAAV-encoding EGFP gene (rAAV(2)-EGFP) in phosphate-buffered saline without and with ultrasound to the retina, respectively. Group D received microbubbles and rAAV(2)-EGFP mixture and ultrasound to the retina. The injection approach was intravitreal injection for all eyes. After 21 days, RGCs were labeled retrogradely with Fluoro-Gold. After 28 days, retinal flat mounts, frozen sections, and pathologic sections were assessed in each group. Expression of EGFP reporter gene was observed on laser confocal microscopy and evaluated according to average optical density and transfected RGC rate. To evaluate adverse effects with retinal flat mounts, labeled RGCs were counted, and retinal pathologic sections were examined. RESULTS When ultrasound parameters (frequency, 0.3 MHz; power, 0.5 W/cm(2); total time, 60 seconds [irradiation time, 5 seconds; interval time, 10 seconds; four times]) were selected, EGFP expression was stronger, and retinas were not damaged. In the second part of the experiment, RGCs were labeled with Fluoro-Gold successfully. Green fluorescence can be observed in labeled RGCs in groups B to D. While average optical density and transfected RGC rate in group D were the highest compared to the other groups, no significant reduction in RGC number was detected with retrograde labeling. No obvious damage was observed with pathologic sections. CONCLUSIONS Ultrasound-mediated microbubble destruction can effectively and safely enhance rAAV delivery to RGCs in rats, and it may represent a novel gene delivery method in gene therapy for glaucomatous optic neuroprotection.
Collapse
Affiliation(s)
- Wenyue Xie
- Department of Ophthalmology, Second Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing 400010, China
| | | | | | | | | | | |
Collapse
|
28
|
Cao H, Zhang GR, Geller AI. Antibody-mediated targeted gene transfer to NMDA NR1-containing neurons in rat neocortex by helper virus-free HSV-1 vector particles containing a chimeric HSV-1 glycoprotein C-staphylococcus A protein. Brain Res 2010; 1351:1-12. [PMID: 20599821 PMCID: PMC2929402 DOI: 10.1016/j.brainres.2010.06.045] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Revised: 06/17/2010] [Accepted: 06/18/2010] [Indexed: 11/16/2022]
Abstract
Because of the heterogeneous cellular composition of the brain, and especially the forebrain, cell type-specific expression will benefit many potential applications of direct gene transfer. The two prevalent approaches for achieving cell type-specific expression are using a cell type-specific promoter or targeting gene transfer to a specific cell type. Targeted gene transfer with Herpes Simplex Virus (HSV-1) vectors modifies glycoprotein C (gC) to replace the heparin binding domain, which binds to many cell types, with a binding activity for a specific cell surface protein. We previously reported targeted gene transfer to nigrostriatal neurons using chimeric gC-glial cell line-derived neurotrophic factor or gC-brain-derived neurotrophic factor protein. Unfortunately, this approach is limited to cells that express the cognate receptor for either neurotrophic factor. Thus, a general strategy for targeting gene transfer to many different types of neurons is desirable. Antibody-mediated targeted gene transfer has been developed for targeting specific virus vectors to specific peripheral cell types; a specific vector particle protein is modified to contain the Staphylococcus A protein ZZ domain, which binds immunoglobulin (Ig) G. Here, we report antibody-mediated targeted gene transfer of HSV-1 vectors to a specific type of forebrain neuron. We constructed a chimeric gC-ZZ protein, and showed this protein is incorporated into vector particles and binds Ig G. Complexes of these vector particles and an antibody to the NMDA receptor NR1 subunit supported targeted gene transfer to NR1-containing neocortical neurons in the rat brain, with long-term (2 months) expression.
Collapse
Affiliation(s)
- Haiyan Cao
- Department of Neurology, West Roxbury VA Hospital/Harvard Medical School, W. Roxbury, MA 02132, USA
| | - Guo-Rong Zhang
- Department of Neurology, West Roxbury VA Hospital/Harvard Medical School, W. Roxbury, MA 02132, USA
| | - Alfred I Geller
- Department of Neurology, West Roxbury VA Hospital/Harvard Medical School, W. Roxbury, MA 02132, USA.
| |
Collapse
|
29
|
Heart-targeted adeno-associated viral vectors selected by in vivo biopanning of a random viral display peptide library. Gene Ther 2010; 17:980-90. [DOI: 10.1038/gt.2010.44] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
30
|
Maguire CA, Gianni D, Meijer DH, Shaket LA, Wakimoto H, Rabkin SD, Gao G, Sena-Esteves M. Directed evolution of adeno-associated virus for glioma cell transduction. J Neurooncol 2010; 96:337-47. [PMID: 19618115 PMCID: PMC2892971 DOI: 10.1007/s11060-009-9972-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2009] [Accepted: 07/06/2009] [Indexed: 10/20/2022]
Abstract
Glioblastoma multiforme (GBM) is a serious form of brain cancer for which there is currently no effective treatment. Alternative strategies such as adeno-associated virus (AAV) vector mediated-genetic modification of brain tumor cells with genes encoding anti-tumor proteins have shown promising results in preclinical models of GBM, although the transduction efficiency of these tumors is often low. As higher transduction efficiency of tumor cells should lead to enhanced therapeutic efficacy, a means to rapidly engineer AAV vectors with improved transduction efficiency for individual tumors is an attractive strategy. Here we tested the possibility of identifying high-efficiency AAV vectors for human U87 glioma cells by selection in culture of a newly constructed chimeric AAV capsid library generated by DNA shuffling of six different AAV cap genes (AAV1, AAV2, AAV5, AAVrh.8, AAV9, AAVrh.10). After seven rounds of selection, we obtained a chimeric AAV capsid that transduces U87 cells at high efficiency (97% at a dose of 10(4) genome copies/cell), and at low doses it was 1.45-1.6-fold better than AAV2, which proved to be the most efficient parental capsid. Interestingly, the new AAV capsid displayed robust gene delivery properties to all glioma cells tested (including primary glioma cells) with relative fluorescence indices ranging from 1- to 14-fold higher than AAV2. The selected vector should be useful for in vitro glioma research when efficient transduction of several cell lines is required, and provides proof-of-concept that an AAV library can be used to generate AAV vectors with enhanced transduction efficiency of glioma cells.
Collapse
Affiliation(s)
- Casey A Maguire
- Department of Neurology, Massachusetts General Hospital, and Neuroscience Program, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Photochemical internalization: a new tool for gene and oligonucleotide delivery. Top Curr Chem (Cham) 2010; 296:251-81. [PMID: 21504105 DOI: 10.1007/128_2010_63] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Photochemical internalization (PCI) is a novel technology for release of endocytosed macromolecules into the cytosol. The technology is based on the use of photosensitizers located in endocytic vesicles. Upon activation by light such photosensitizers induce a release of macromolecules from their compartmentalization in endocytic vesicles. PCI has been shown to increase the biological activity of a large variety of macromolecules and other molecules that do not readily penetrate the plasma membrane, including type I ribosome-inactivating proteins, immunotoxins, plasmids, adenovirus, various oligonucleotides, dendrimer-based delivery of chemotherapeutica and unconjugated chemotherapeutica such as bleomycin and doxorubicin. This review will present the basis for the PCI concept and the most recent significant developments.
Collapse
|
32
|
Adeno-associated viral vector serotypes 1 and 5 targeted to the neonatal rat and pig striatum induce widespread transgene expression in the forebrain. Exp Neurol 2009; 222:70-85. [PMID: 20025873 DOI: 10.1016/j.expneurol.2009.12.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Revised: 12/08/2009] [Accepted: 12/09/2009] [Indexed: 11/24/2022]
Abstract
Viral vector-mediated gene transfer has emerged as a powerful means to target transgene expression in the central nervous system. Here we characterized the efficacy of serotypes 1 and 5 recombinant adeno-associated virus (rAAV) vectors encoding green fluorescent protein (GFP) after stereotaxic delivery to the neonatal rat and minipig striatum. The efficiency of GFP expression and the phenotype of GFP-positive cells were assessed within the forebrain at different time points up to 12 months after surgery. Both rAAV1-GFP and rAAV5-GFP delivery resulted in transduction of the striatum as well as striatal input and output areas, including large parts of the cortex. In both species, rAAV5 resulted in a more widespread transgene expression compared to rAAV1. In neonatal rats, rAAV5 also transduced several other areas such as the olfactory bulbs, hippocampus, and septum. Phenotypic analysis of the GFP-positive cells, performed using immunohistochemistry and confocal microscopy, showed that most of the GFP-positive cells by either serotype were NeuN-positive neuronal profiles. The rAAV5 vector further displayed the ability to transduce non-neuronal cell types in both rats and pigs, albeit at a low frequency. Our results show that striatal delivery of rAAV5 vectors in the neonatal brain represents a useful tool to express genes of interest both in the basal ganglia and the neocortex. Furthermore, we apply, for the first time, viral vector-mediated gene transfer to the pig brain providing the opportunity to study effects of genetic manipulation in this non-primate large animal species. Finally, we generated an atlas of the Göttingen minipig brain for guiding future studies in this large animal species.
Collapse
|
33
|
White AF, Mazur M, Sorscher EJ, Zinn KR, Ponnazhagan S. Genetic modification of adeno-associated viral vector type 2 capsid enhances gene transfer efficiency in polarized human airway epithelial cells. Hum Gene Ther 2009; 19:1407-14. [PMID: 18778196 DOI: 10.1089/hum.2008.117] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Cystic fibrosis (CF) is a common genetic disease characterized by defects in the expression of the CF transmembrane conductance regulator (CFTR) gene. Gene therapy offers better hope for the treatment of CF. Adeno-associated viral (AAV) vectors are capable of stable expression with low immunogenicity. Despite their potential in CF gene therapy, gene transfer efficiency by AAV is limited because of pathophysiological barriers in these patients. Although a few AAV serotypes have shown better transduction compared with the AAV2-based vectors, gene transfer efficiency in human airway epithelium has still not reached therapeutic levels. To engineer better AAV vectors for enhanced gene delivery in human airway epithelium, we developed and characterized mutant AAV vectors by genetic capsid modification, modeling the well-characterized AAV2 serotype. We genetically incorporated putative high-affinity peptide ligands to human airway epithelium on the GH loop region of AAV2 capsid protein. Six independent mutant AAV were constructed, containing peptide ligands previously reported to bind with high affinity for known and unknown receptors on human airway epithelial cells. The vectors were tested on nonairway cells and nonpolarized and polarized human airway epithelial cells for enhanced infectivity. One of the mutant vectors, with the peptide sequence THALWHT, not only showed the highest transduction in undifferentiated human airway epithelial cells but also indicated significant transduction in polarized cells. Interestingly, this modified vector was also able to infect cells independently of the heparan sulfate proteoglycan receptor. Incorporation of this ligand on other AAV serotypes, which have shown improved gene transfer efficiency in the human airway epithelium, may enhance the application of AAV vectors in CF gene therapy.
Collapse
Affiliation(s)
- April F White
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | |
Collapse
|
34
|
Abstract
Cardiac gene and cell therapy have both entered clinical trials aimed at ameliorating ventricular dysfunction in patients with chronic congestive heart failure. The transduction of myocardial cells with viral constructs encoding a specific cardiomyocyte Ca(2+) pump in the sarcoplasmic reticulum (SR), SRCa(2+)-ATPase has been shown to correct deficient Ca(2+) handling in cardiomyocytes and improvements in contractility in preclinical studies, thus leading to the first clinical trial of gene therapy for heart failure. In cell therapy, it is not clear whether beneficial effects are cell-type specific and how improvements in contractility are brought about. Despite these uncertainties, a number of clinical trials are under way, supported by safety and efficacy data from trials of cell therapy in the setting of myocardial infarction. Safety concerns for gene therapy center on inflammatory and immune responses triggered by viral constructs, and for cell therapy with myoblast cells, the major concern is increased incidence of ventricular arrhythmia after cell transplantation. Principles and mechanisms of action of gene and cell therapy for heart failure are discussed, together with the potential influence of reactive oxygen species on the efficacy of these treatments and the status of myocardial-delivery techniques for viral constructs and cells.
Collapse
Affiliation(s)
- Ebo D de Muinck
- Departments of Medicine and Physiology, Dartmouth Medical School, Lebanon, New Hampshire 03756, USA.
| |
Collapse
|
35
|
Madsen D, Cantwell ER, O'Brien T, Johnson PA, Mahon BP. Adeno-associated virus serotype 2 induces cell-mediated immune responses directed against multiple epitopes of the capsid protein VP1. J Gen Virol 2009; 90:2622-2633. [PMID: 19641045 PMCID: PMC2885037 DOI: 10.1099/vir.0.014175-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Adeno-associated virus serotype 2 (AAV-2) has been developed as a gene therapy vector. Antibody and cell-mediated immune responses to AAV-2 or AAV-2-transfected cells may confound the therapeutic use of such vectors in clinical practice. In one of the most detailed examinations of AAV-2 immunity in humans to date, cell-mediated and humoral immune responses to AAV-2 were characterized from a panel of healthy blood donors. The extent of AAV-2-specific antibody in humans was determined by examination of circulating AAV-2-specific total IgG levels in plasma from 45 normal donors. Forty-one donors were seropositive and responses were dominated by IgG1 and IgG2 subclasses. Conversely, AAV-2-specific IgG3 levels were consistently low in all donors. Cell-mediated immune recall responses were detectable in nearly half the population studied. In vitro restimulation with AAV-2 of peripheral blood mononuclear cell cultures from 16 donors elicited gamma interferon (IFN-γ) (ten donors), interleukin-10 (IL-10) (eight donors) and interleukin-13 (IL-13) (four donors) responses. Using a series of overlapping peptides derived from the sequence of the VP1 viral capsid protein, a total of 59 candidate T-cell epitopes were identified. Human leukocyte antigen characterization of donors revealed that the population studied included diverse haplotypes, but that at least 17 epitopes were recognized by multiple donors and could be regarded as immunodominant. These data indicate that robust immunological memory to AAV-2 is established. The diversity of sequences recognized suggests that attempts to modify the AAV-2 capsid, as a strategy to avoid confounding immunity, will not be feasible.
Collapse
Affiliation(s)
- Declan Madsen
- Cellular Immunology Laboratory, Institute of Immunology, National University of Ireland Maynooth, County Kildare, Ireland
| | - Emma R. Cantwell
- Cellular Immunology Laboratory, Institute of Immunology, National University of Ireland Maynooth, County Kildare, Ireland
| | - Timothy O'Brien
- Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Galway, Ireland
| | - Patricia A. Johnson
- Viral Immunology Laboratory, School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Bernard P. Mahon
- Cellular Immunology Laboratory, Institute of Immunology, National University of Ireland Maynooth, County Kildare, Ireland
| |
Collapse
|
36
|
Kasuya T, Kuroda S. Nanoparticles for human liver-specific drug and gene delivery systems: in vitro and in vivo advances. Expert Opin Drug Deliv 2009; 6:39-52. [PMID: 19236207 DOI: 10.1517/17425240802622096] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A wide variety of nanoparticles (NPs) that can deliver incorporated therapeutic materials such as compounds, proteins, genes and siRNAs to the human liver have been developed to treat liver-related diseases. This review describes NP-based drug and gene delivery systems such as liposomes (including lipoplex), polymer micelles, polymers (including polyplex) and viral vectors. It focuses upon the modification of these NPs to enhance liver specificity or delivery efficiency in vitro and in vivo. We discuss recent advances in drug and gene delivery systems specific to the human liver utilizing bio-nanocapsules comprising hepatitis B virus (HBV) envelope L protein, which has a pivotal role in HBV infection. These NP-based medicines may offer novel strategies for the treatment of liver-related diseases and contribute to the development of nanomedicines targeting other tissues.
Collapse
Affiliation(s)
- Takeshi Kasuya
- Osaka University, Institute of Scientific and Industrial Research, Department of Structural Molecular Biology, Ibaraki, Japan
| | | |
Collapse
|
37
|
Driessen WHP, Ozawa MG, Arap W, Pasqualini R. Ligand-directed cancer gene therapy to angiogenic vasculature. ADVANCES IN GENETICS 2009; 67:103-121. [PMID: 19914451 PMCID: PMC7172741 DOI: 10.1016/s0065-2660(09)67004-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Gene therapy strategies in cancer have remained an active area of preclinical and clinical research. One of the current limitations to successful trials is the relative transduction efficiency to produce a therapeutic effect. While intratumoral injections are the mainstay of many treatment regimens to date, this approach is hindered by hydrostatic pressures within the tumor and is not always applicable to all tumor subtypes. Vascular-targeting strategies introduce an alternative method to deliver vectors with higher local concentrations and minimization of systemic toxicity. Moreover, therapeutic targeting of angiogenic vasculature often leads to enhanced bystander effects, improving efficacy. While identification of functional and systemically accessible molecular targets is challenging, approaches, such as in vivo phage display and phage-based viral delivery vectors, provide a platform upon which vascular targeting of vectors may become a viable and translational approach.
Collapse
Affiliation(s)
- Wouter H P Driessen
- David H. Koch Center, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - Michael G Ozawa
- David H. Koch Center, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - Wadih Arap
- David H. Koch Center, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - Renata Pasqualini
- David H. Koch Center, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
38
|
Abstract
A number of preclinical studies have shown the adeno-associated virus (AAV) to be an efficient vehicle for gene therapy. Clinical studies successfully demonstrated its potential for in vivo gene transfer. The complexity of host-vector interactions when progressing from small to large animal models, and eventually to humans, has impeded translation of AAV technology to the clinic. One approach to address this complexity has been to explore the biological characteristics of variations in AAV capsid structure. Initial strategies characterized the naturally occurring capsid variants from mammalian species. The structural and functional knowledge gathered on these natural AAV variants as vectors has led to the first series of second-generation vectors that aim at specifically improving certain properties by rational design of the capsid. A third exciting approach uses directed evolution to isolate vectors that are able to overcome selective pressures applied in the laboratory and thereby steer the capsid to evolve toward improved functionality.
Collapse
|
39
|
Capsid modification of adeno-associated virus and tumor targeting gene therapy. Sci Bull (Beijing) 2008. [DOI: 10.1007/s11434-008-0487-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
40
|
Federici T, Liu JK, Teng Q, Garrity-Moses M, Yang J, Boulis NM. Neuronal affinity of a C7C loop peptide identified through phage display. J Drug Target 2008; 14:263-71. [PMID: 16882546 DOI: 10.1080/10611860600763079] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Phage display is a promising tool for the screening of peptides with high affinity for specific cells. Here we describe a novel peptide with neuronal affinity isolated from a C7C library. We designed a two-tiered biopanning strategy initially selecting for ganglioside binding and subsequently selecting for binding to PC12 cells. At the completion of biopanning, 54.8% of phage clones bore the identical peptide (Tet.C7C.1). Immunofluorescence confirmed selective binding of this clone to differentiated PC12 cells. Tet.C7C.1 was synthesized and fluorescein conjugated. The synthetic peptide binds neuronal cell lines (SH-SY5Y, NSC-34 and PC12 cells) and tissue (DRG and spinal cord). The C7C structure creates a loop that minimizes the impact of peptide insertion on the confirmation of the recipient protein. Small loop peptides have the ideal characteristics for modification of viral vector capsids without undermining genome packaging. The neuronal binding properties of this peptide may be applied in the development of neurotropic viral vectors.
Collapse
Affiliation(s)
- Thais Federici
- Department of Neuroscience and Center for Neurological Restoration, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH, 44195, USA.
| | | | | | | | | | | |
Collapse
|
41
|
Abstract
This unit is devoted to safety issues that must be considered when generating and working with vectors for gene therapy. A short description of the relevant regulatory bodies and mechanisms is followed by a general discussion of containment and safety procedures. This is followed by vector-specific guidelines for nine commonly used vector systems, including adenovirus, vaccinia, HSV-1, lentivirus, and nonviral vectors. Recent tragic experiences with retroviral integration near oncogene loci demonstrate the surprise effects of successful gene therapy. The unit also outlines the problems encountered when taking a promising vector to clinical trials.
Collapse
Affiliation(s)
- Andrew Braun
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
42
|
In vitro and in vivo gene therapy vector evolution via multispecies interbreeding and retargeting of adeno-associated viruses. J Virol 2008; 82:5887-911. [PMID: 18400866 DOI: 10.1128/jvi.00254-08] [Citation(s) in RCA: 531] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Adeno-associated virus (AAV) serotypes differ broadly in transduction efficacies and tissue tropisms and thus hold enormous potential as vectors for human gene therapy. In reality, however, their use in patients is restricted by prevalent anti-AAV immunity or by their inadequate performance in specific targets, exemplified by the AAV type 2 (AAV-2) prototype in the liver. Here, we attempted to merge desirable qualities of multiple natural AAV isolates by an adapted DNA family shuffling technology to create a complex library of hybrid capsids from eight different wild-type viruses. Selection on primary or transformed human hepatocytes yielded pools of hybrids from five of the starting serotypes: 2, 4, 5, 8, and 9. More stringent selection with pooled human antisera (intravenous immunoglobulin [IVIG]) then led to the selection of a single type 2/type 8/type 9 chimera, AAV-DJ, distinguished from its closest natural relative (AAV-2) by 60 capsid amino acids. Recombinant AAV-DJ vectors outperformed eight standard AAV serotypes in culture and greatly surpassed AAV-2 in livers of naïve and IVIG-immunized mice. A heparin binding domain in AAV-DJ was found to limit biodistribution to the liver (and a few other tissues) and to affect vector dose response and antibody neutralization. Moreover, we report the first successful in vivo biopanning of AAV capsids by using a new AAV-DJ-derived viral peptide display library. Two peptides enriched after serial passaging in mouse lungs mediated the retargeting of AAV-DJ vectors to distinct alveolar cells. Our study validates DNA family shuffling and viral peptide display as two powerful and compatible approaches to the molecular evolution of novel AAV vectors for human gene therapy applications.
Collapse
|
43
|
Augmented transgene expression in transformed cells using a parvoviral hybrid vector. Cancer Gene Ther 2008; 15:252-67. [PMID: 18202715 DOI: 10.1038/sj.cgt.7701113] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Autonomous parvoviruses possess an intrinsic oncotropism based on viral genetic elements controlling gene expression and genome replication. We constructed a hybrid vector consisting of the H1 parvovirus-derived expression cassette comprising the p4 promoter, the ns1 gene and the p38 promoter flanked by the adeno-associated viruses 2 (AAV2) inverted terminal repeats and packaged into AAV2 capsids. Gene transduction using this vector could be stimulated by coinfection with adenovirus, by irradiation or treatment with genotoxic agents, similar to standard AAV2 vectors. However, the latter were in most cases less efficient in gene transduction than the hybrid vector. With the new vector, tumor cell-selective increase in transgene expression was observed in pairs of transformed and non-transformed cells, leading to selective killing of the transformed cells after expression of a prodrug-converting enzyme. Preferential gene expression in tumor versus normal liver tissue was also observed in vivo in a syngeneic rat model. Comparative transduction of a panel of different tumor cell lines with the H1 and the H1/AAV hybrid vector showed a preference of each vector for distinct cell types, probably reflecting the dependence of the viral tropism on capsid determinants.
Collapse
|
44
|
Driessen WHP, Fujii N, Tamamura H, Sullivan SM. Development of peptide-targeted lipoplexes to CXCR4-expressing rat glioma cells and rat proliferating endothelial cells. Mol Ther 2008; 16:516-24. [PMID: 18195720 DOI: 10.1038/sj.mt.6300388] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
A peptide analog, 4-fluorobenzoyl-RR-(L-3-(2-naphthyl)alanine)-CYEK-(L-citrulline)-PYR-(L-citrulline)-CR, covalently linked to a phospholipid, was used for targeting a lipid-based gene delivery vehicle to CXCR4(+)-cells. Characterization of transfection activity was done in vitro using a transformed rat glioma cell line (RG2) that expresses CXCR4. The substitution of the targeting lipid at increasing mole percentages in the place of helper lipids yielded a progressive increase in reporter gene expression, reaching a maximum of 2.5 times the control value at 20 mol% of ligand. The substitution of helper lipids with cysteine-derivatized phospholipid analog or phosphatidylethanolamine resulted in a progressive decrease in transfection activity, with complete inactivation of the complex occurring at 20 mol%. A DNA dose-response with 10 mol% of lipopeptide reduced the effective DNA dose at least fivefold with regard to the number of transfected cells and >20-fold with regard to the amount of gene expression. Gene transfer to rat endothelial cells was studied in the context of an arterial organ culture. Mesenteric arteries were cannulated and maintained in culture for up to 4 days. CXCR4 cell-surface expression on endothelial cells was induced after overnight incubation with vascular endothelial growth factor (VEGF). Gene transfer studies showed that only the peptide-targeted lipoplexes transfected the endothelium, and only after CXCR4 had been induced with VEGF. These results demonstrate that non-viral transfection complexes can be targeted to cells expressing CXCR4, and that gene transfer is dependent upon cell surface receptor expression levels.
Collapse
Affiliation(s)
- Wouter H P Driessen
- Department of Pharmaceutics Sciences, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | | | | | | |
Collapse
|
45
|
Poller W, Suckau L, Pinkert S, Fechner H. RNA Interference and MicroRNA Modulation for the Treatment of Cardiac Disorders. RNA TECHNOLOGIES IN CARDIOVASCULAR MEDICINE AND RESEARCH 2008. [PMCID: PMC7121055 DOI: 10.1007/978-3-540-78709-9_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The current status and challenges of RNA interference (RNAi) and microRNA modulation strategies for the treatment of myocardial disorders are discussed and related to the classical gene therapeutic approaches of the past decade. Section 2 summarizes the key issues of current vector technologies which determine if they may be suitable for clinical translation of experimental RNAi or microRNA therapeutic protocols. We then present and discuss examples dealing with the potential of cardiac RNAi therapy. First, an approach to block a key early step in the pathogenesis of a virus-induced cardiomyopathy by RNAi targeting of a cellular receptor for cardiopathogenic viruses (Section 3). Second, an approach to improve cardiac function by RNAi targeting of late pathway of heart failure pathogenesis common to myocardial disorders of multiple etiologies. This strategy is directed at myocardial Ca2+ homeostasis which is disturbed in heart failure due to coronary heart disease, heart valve dysfunction, cardiac inflammation, or genetic defects (Section 4). Whereas the first type of strategies (directed at early pathogenesis) need to be tailor-made for each different type of pathomechanism, the second type (targeting late common pathways) has a much broader range of application. This advantage of the second type of approaches is of key importance since enormous efforts need to be undertaken before any regulatory RNA therapy enters the stage of possible clinical translation. If then the number of patients eligible for this protocol is large, the actual transformation of the experimental therapy into a new therapeutic option of clinical importance is far more likely to occur.
Collapse
|
46
|
Michelfelder S, Lee MK, deLima-Hahn E, Wilmes T, Kaul F, Müller O, Kleinschmidt JA, Trepel M. Vectors selected from adeno-associated viral display peptide libraries for leukemia cell-targeted cytotoxic gene therapy. Exp Hematol 2007; 35:1766-76. [PMID: 17920758 DOI: 10.1016/j.exphem.2007.07.018] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2007] [Revised: 07/17/2007] [Accepted: 07/23/2007] [Indexed: 01/02/2023]
Abstract
OBJECTIVE For acute myeloid leukemia (AML), gene therapy may be used to treat patients refractory to conventional chemotherapy. However, availability of vectors sufficiently and specifically transducing this cell type is very limited. METHOD Here we report the selection of capsid-modified adeno-associated viral (AAV) vectors targeting Kasumi-1 AML cells by screening random AAV displayed peptide libraries. RESULTS The peptide inserts of the enriched capsid mutants share a common sequence motif. The same motif was selected in an independent library screening on HL-60 AML cells. Recombinant targeted vectors displaying the selected peptides transduced the target leukemia cells they have been selected on up to 500-fold more efficiently compared to AAV vectors with control peptide inserts. One of the selected clones (NQVGSWS) also efficiently transduced all members of a panel of four other AML cell lines. Binding and blocking experiments showed that NQVGSWS binding to leukemia cells is independent of the wild-type AAV-2 receptor heparin sulfate proteoglycan. Transduction assays on a panel of hematopoietic and nonhematopoietic cell lines showed that the NQVGSWS capsid was able to overcome resistance to AAV transduction, especially in hematopoietic cancer cells, whereas normal peripheral blood mononuclear cells and CD34(+) hematopoietic progenitor cells were not transduced. CONCLUSIONS Consequently, recombinant targeted NQVGSWS AAV vectors harboring a suicide gene conferred selective killing to Kasumi-1 cells, but not to control cells. This suggests that the AAV mutant selected here may be used as a tool to target therapeutic genes to AML cells.
Collapse
Affiliation(s)
- Stefan Michelfelder
- Department of Hematology and Oncology, University of Freiburg Medical Center, Freiburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Kwon I, Schaffer DV. Designer gene delivery vectors: molecular engineering and evolution of adeno-associated viral vectors for enhanced gene transfer. Pharm Res 2007; 25:489-99. [PMID: 17763830 PMCID: PMC2265771 DOI: 10.1007/s11095-007-9431-0] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2007] [Accepted: 08/03/2007] [Indexed: 12/23/2022]
Abstract
Gene delivery vectors based on adeno-associated virus (AAV) are highly promising due to several desirable features of this parent virus, including a lack of pathogenicity, efficient infection of dividing and non-dividing cells, and sustained maintenance of the viral genome. However, several problems should be addressed to enhance the utility of AAV vectors, particularly those based on AAV2, the best characterized AAV serotype. First, altering viral tropism would be advantageous for broadening its utility in various tissue or cell types. In response to this need, vector pseudotyping, mosaic capsids, and targeting ligand insertion into the capsid have shown promise for altering AAV specificity. In addition, library selection and directed evolution have recently emerged as promising approaches to modulate AAV tropism despite limited knowledge of viral structure-function relationships. Second, pre-existing immunity to AAV must be addressed for successful clinical application of AAV vectors. "Shielding" polymers, site-directed mutagenesis, and alternative AAV serotypes have shown success in avoiding immune neutralization. Furthermore, directed evolution of the AAV capsid is a high throughput approach that has yielded vectors with substantial resistance to neutralizing antibodies. Molecular engineering and directed evolution of AAV vectors therefore offer promise for generating 'designer' gene delivery vectors with enhanced properties.
Collapse
Affiliation(s)
- Inchan Kwon
- Department of Chemical Engineering and Helen Wills Neuroscience Institute, University of California, 201 Gilman Hall, Berkeley, California 94720-1462 USA
| | - David V. Schaffer
- Department of Chemical Engineering and Helen Wills Neuroscience Institute, University of California, 201 Gilman Hall, Berkeley, California 94720-1462 USA
| |
Collapse
|
48
|
Guan YS, La Z, Yang L, He Q, Li P. p53 gene in treatment of hepatic carcinoma: status quo. World J Gastroenterol 2007; 13:985-992. [PMID: 17373730 PMCID: PMC4146884 DOI: 10.3748/wjg.v13.i7.985] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2006] [Revised: 12/12/2006] [Accepted: 01/16/2007] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the 10 most common cancers worldwide. There is no ideal treatment for HCC yet and many researchers are trying to improve the effects of treatment by changing therapeutic strategies. As the majority of human cancers seem to exhibit either abnormal p53 gene or disrupted p53 gene activation pathways, intervention to restore wild-type p53 (wt-p53) activities is an attractive anti-cancer therapy including HCC. Abnormalities of p53 are also considered a predisposition factor for hepatocarcinogenesis. p53 is frequently mutated in HCC. Most HCCs have defects in the p53-mediated apoptotic pathway although they carry wt-p53. High expression of p53 in vivo may exert therapeutic effects on HCC in two aspects: (1) High expression of exogenous p53 protein induces apoptosis of tumor cells by inhibiting proliferation of cells through several biologic pathways and (2) Exogenous p53 renders HCC more sensitive to some chemotherapeutic agents. Several approaches have been designed for the treatment of HCC via the p53 pathway by restoring the tumor suppression function from inactivation, rescuing the mutated p53 gene from instability, or delivering therapeutic exogenous p53. Products with p53 status as the target have been studied extensively in vitro and in vivo. This review elaborates some therapeutic mechanisms and advances in using recombinant human adenovirus p53 and oncolytic virus products for the treatment of HCC.
Collapse
Affiliation(s)
- Yong-Song Guan
- Department of Radiology and Oncology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China.
| | | | | | | | | |
Collapse
|
49
|
Waterkamp DA, Müller OJ, Ying Y, Trepel M, Kleinschmidt JA. Isolation of targeted AAV2 vectors from novel virus display libraries. J Gene Med 2007; 8:1307-19. [PMID: 16955542 DOI: 10.1002/jgm.967] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Random peptide ligands displayed on viral capsids are emerging tools for selection of targeted gene transfer vectors even without prior knowledge of the potential target cell receptor. We have previously introduced adeno-associated viral (AAV)-displayed peptide libraries that ensure encoding of displayed peptides by the packaged AAV genome. A major limitation of these libraries is their contamination with wild-type (wt) AAV. Here we describe a novel and improved library production system that reliably avoids generation of wt AAV by use of a synthetic cap gene. Selection of targeted AAV vectors from wt-containing and the novel wt-free libraries on cell types with different permissivity for wt AAV2 replication suggested the superiority of the wt-free library. However, from both libraries highly specific peptide sequence motifs were selected which improved transduction of cells with moderate or low permissivity for AAV2 replication. Strong reduction of HeLa cell transduction compared to wt AAV2 and only low level transduction of non-target cells by some selected clones showed that not only the efficiency but also the specificity of gene transfer was improved. In conclusion, our study validates and improves the unique potential of virus display libraries for the development of targeted gene transfer vectors.
Collapse
Affiliation(s)
- Daniel A Waterkamp
- Deutsches Krebsforschungszentrum, Tumor Virologie, Im Neuenheimer Feld 242, D-69120 Heidelberg, Germany
| | | | | | | | | |
Collapse
|
50
|
Berg K, Høgset A, Prasmickaite L, Weyergang A, Bonsted A, Dietze A, Lou PJ, Bown S, Norum OJ, Møllergård HMT, Selbo PK. Photochemical internalization (PCI): A novel technology for activation of endocytosed therapeutic agents. ACTA ACUST UNITED AC 2006. [DOI: 10.1016/j.mla.2006.08.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|