1
|
Cornetta K, Lin TY, Tao H, Huang J, Piskorowski J, Wilcox P, Xie D, Yao J, House K, Nance E, Duffy L. Product Enhanced Reverse Transcriptase for assessing replication competent virus in vectors retroviral vectors pseudotyped with GALV and VSV-G envelopes. J Virol Methods 2024; 329:114988. [PMID: 38908550 PMCID: PMC11368651 DOI: 10.1016/j.jviromet.2024.114988] [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: 02/17/2024] [Revised: 06/02/2024] [Accepted: 06/16/2024] [Indexed: 06/24/2024]
Abstract
We evaluated the use of the Product Enhanced Reverse Transcriptase (PERT) assay as a means of detecting virus in retroviral vectors products pseudotyped with Gibbon Ape Leukemia Virus (GALV) and Vesicular Stomatitis Virus G (VSVG) envelopes. PERT provides greater standardization than the S+/L- assay which has been used extensively in virus detection. A challenge is that PERT will also detect residual retroviral vectors as vector particles contain reverse transcriptase. Vector products were cultured for 3 weeks on HEK293 cells to amplify any potential virus. In addition, vector supernatant and end-of-production cells were spiked with GALV to evaluate for inhibition by the test article. Results of PERT and the S+/L- assay were compared. PERT and S+/L- assays were both effective in detecting virus. Vector supernatants were negative at the end of 3 weeks of culture by PERT for both GAVL and VSVG pseudotyped vector. In contrast, end-of-production cells were positive by PERT due to persistent vector producing cells. A one-week culture of cell-free media obtained at the 3 weeks timepoint allowed distinction of virus-free test articles from those with virus. The PERT assay is suitable for detecting replication competent retrovirus in vector products pseudotyped with GALV and VSVG envelopes.
Collapse
Affiliation(s)
- Kenneth Cornetta
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, USA.
| | - Tsai-Yu Lin
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, USA
| | - Haipeng Tao
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Jianping Huang
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Jordon Piskorowski
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, USA
| | - Parker Wilcox
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, USA
| | - Danhui Xie
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, USA
| | - Jing Yao
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, USA
| | - Kimberley House
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, USA
| | - Emily Nance
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, USA
| | - Lisa Duffy
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, USA
| |
Collapse
|
2
|
van Heuvel Y, Schatz S, Hein M, Dogra T, Kazenmaier D, Tschorn N, Genzel Y, Stitz J. Novel suspension retroviral packaging cells generated by transposition using transposase encoding mRNA advance vector yields and enable production in bioreactors. Front Bioeng Biotechnol 2023; 11:1076524. [PMID: 37082212 PMCID: PMC10112512 DOI: 10.3389/fbioe.2023.1076524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 03/24/2023] [Indexed: 04/22/2023] Open
Abstract
To date, the establishment of high-titer stable viral packaging cells (VPCs) at large scale for gene therapeutic applications is very time- and cost-intensive. Here we report the establishment of three human suspension 293-F-derived ecotropic MLV-based VPCs. The classic stable transfection of an EGFP-expressing transfer vector resulted in a polyclonal VPC pool that facilitated cultivation in shake flasks of 100 mL volumes and yielded high functional titers of more than 1 × 106 transducing units/mL (TU/mL). When the transfer vector was flanked by transposon terminal inverted repeats (TIRs) and upon co-transfection of a plasmid encoding for the transposase, productivities could be slightly elevated to more than 3 × 106 TU/mL. In contrast and using mRNA encoding for the transposase, as a proof of concept, productivities were drastically improved by more than ten-fold exceeding 5 × 107 TU/mL. In addition, these VPC pools were generated within only 3 weeks. The production volume was successfully scaled up to 500 mL employing a stirred-tank bioreactor (STR). We anticipate that the stable transposition of transfer vectors employing transposase transcripts will be of utility for the future establishment of high-yield VPCs producing pseudotype vector particles with a broader host tropism on a large scale.
Collapse
Affiliation(s)
- Yasemin van Heuvel
- Research Group Medical Biotechnology and Bioengineering, Faculty of Applied Natural Sciences, University of Applied Sciences Cologne, Campus Leverkusen, Cologne, Germany
- Institute of Technical Chemistry, Gottfried Wilhelm Leibniz University Hannover, Hanover, Germany
| | - Stefanie Schatz
- Research Group Medical Biotechnology and Bioengineering, Faculty of Applied Natural Sciences, University of Applied Sciences Cologne, Campus Leverkusen, Cologne, Germany
- Institute of Technical Chemistry, Gottfried Wilhelm Leibniz University Hannover, Hanover, Germany
| | - Marc Hein
- Chair of Bioprocess Engineering, Otto-Von-Guericke-University Magdeburg, Magdeburg, Germany
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Tanya Dogra
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Daniel Kazenmaier
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
- Faculty of Biotechnology, University of Applied Sciences Mannheim, Mannheim, Germany
| | - Natalie Tschorn
- Research Group Medical Biotechnology and Bioengineering, Faculty of Applied Natural Sciences, University of Applied Sciences Cologne, Campus Leverkusen, Cologne, Germany
- Institute of Technical Chemistry, Gottfried Wilhelm Leibniz University Hannover, Hanover, Germany
| | - Yvonne Genzel
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Jörn Stitz
- Research Group Medical Biotechnology and Bioengineering, Faculty of Applied Natural Sciences, University of Applied Sciences Cologne, Campus Leverkusen, Cologne, Germany
- *Correspondence: Jörn Stitz,
| |
Collapse
|
3
|
Inadvertent Transfer of Murine VL30 Retrotransposons to CAR-T Cells. ADVANCES IN CELL AND GENE THERAPY 2022; 2022. [PMID: 36081760 PMCID: PMC9450689 DOI: 10.1155/2022/6435077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
For more than a decade, genetically engineered autologous T-cells have been successfully employed as immunotherapy drugs for patients with incurable blood cancers. The active components in some of these game-changing medicines are autologous T-cells that express viral vector-delivered chimeric antigen receptors (CARs), which specifically target proteins that are preferentially expressed on cancer cells. Some of these therapeutic CAR expressing T-cells (CAR-Ts) are engineered via transduction with
-retroviral vectors (
-RVVs) produced in a stable producer cell line that was derived from murine PG13 packaging cells (ATCC CRL-10686). Earlier studies reported on the copackaging of murine virus-like 30S RNA (VL30) genomes with
-retroviral vectors generated in murine stable packaging cells. In an earlier study, VL30 mRNA was found to enhance the metastatic potential of human melanoma cells. These findings raise biosafety concerns regarding the possibility that therapeutic CAR-Ts have been inadvertently contaminated with potentially oncogenic VL30 retrotransposons. In this study, we demonstrated the presence of infectious VL30 particles in PG13 cell-conditioned media and observed the ability of these particles to deliver transcriptionally active VL30 genomes to human cells. Notably, VL30 genomes packaged by HIV-1-based vector particles transduced naïve human cells in culture. Furthermore, we detected the transfer and expression of VL30 genomes in clinical-grade CAR-T cells generated by transduction with PG13 cell-derived
-retroviral vectors. Our findings raise biosafety concerns regarding the use of murine packaging cell lines in ongoing clinical applications.
Collapse
|
4
|
Alyami EM, Tarar A, Peng CA. Less phagocytosis of viral vectors by tethering with CD47 ectodomain. J Mater Chem B 2021; 10:64-77. [PMID: 34846059 DOI: 10.1039/d1tb01815a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Many viral vectors, which are effective when administrated in situ, lack efficacy when delivered intravenously. The key reason for this is the rapid clearance of the viruses from the blood circulation via the immune system before they reach target sites. Therefore, avoiding their clearance by the immune system is essential. In this study, lentiviral vectors were tethered with the ectodomain of self-marker protein CD47 to suppress phagocytosis via interacting with SIRPα on the outer membrane of macrophage cells. CD47 ectodomain and core-streptavidin fusion gene (CD47ED-coreSA) was constructed into pET-30a(+) plasmid and transformed into Lemo21 (DE3) competent E. coli cells. The expressed CD47ED-coreSA chimeric protein was purified by cobalt-nitrilotriacetate affinity column and characterized by SDS-PAGE and western blot. The purified chimeric protein was anchored on biotinylated lentivirus via biotin-streptavidin binding. The CD47ED-capped lentiviruses encoding GFP were used to infect J774A.1 macrophage cells to assess the impact on phagocytosis. Our results showed that the overexpressed CD47ED-coreSA chimeric protein was purified and bound on the surface of biotinylated lentivirus which was confirmed via immunoblotting assay. The process to produce biotinylated lentivirus did not affect native viral infectivity. It was shown that the level of GFP expression in J774A.1 macrophages transduced with CD47ED-lentiviruses was threefold lower in comparison to control lentiviruses, indicating an antiphagocytic effect triggered by the interaction of CD47ED and SIRPα. Through the test of blocking antibodies against CD47ED and/or SIRPα, it was confirmed that the phagocytosis inhibition was mediated through the CD47ED-SIRPα axis signaling. In conclusion, surface immobilization of CD47ED on lentiviral vectors inhibits their phagocytosis by macrophages. The chimeric protein of CD47 ectodomain and core-streptavidin is effective in mediating the surface binding and endowing the lentiviral nanoparticles with the antiphagocytic property.
Collapse
Affiliation(s)
- Esmael M Alyami
- Department of Chemical and Biological Engineering, University of Idaho, Engineering Physics Building 410, 875 Perimeter Drive, Moscow, ID 83844-0904, USA.
| | - Ammar Tarar
- Department of Chemical and Biological Engineering, University of Idaho, Engineering Physics Building 410, 875 Perimeter Drive, Moscow, ID 83844-0904, USA.
| | - Ching-An Peng
- Department of Chemical and Biological Engineering, University of Idaho, Engineering Physics Building 410, 875 Perimeter Drive, Moscow, ID 83844-0904, USA.
| |
Collapse
|
5
|
Raes L, De Smedt SC, Raemdonck K, Braeckmans K. Non-viral transfection technologies for next-generation therapeutic T cell engineering. Biotechnol Adv 2021; 49:107760. [PMID: 33932532 DOI: 10.1016/j.biotechadv.2021.107760] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 04/24/2021] [Accepted: 04/24/2021] [Indexed: 12/24/2022]
Abstract
Genetically engineered T cells have sparked interest in advanced cancer treatment, reaching a milestone in 2017 with two FDA-approvals for CD19-directed chimeric antigen receptor (CAR) T cell therapeutics. It is becoming clear that the next generation of CAR T cell therapies will demand more complex engineering strategies and combinations thereof, including the use of revolutionary gene editing approaches. To date, manufacturing of CAR T cells mostly relies on γ-retroviral or lentiviral vectors, but their use is associated with several drawbacks, including safety issues, high manufacturing cost and vector capacity constraints. Non-viral approaches, including membrane permeabilization and carrier-based techniques, have therefore gained a lot of interest to replace viral transductions in the manufacturing of T cell therapeutics. This review provides an in-depth discussion on the avid search for alternatives to viral vectors, discusses key considerations for T cell engineering technologies, and provides an overview of the emerging spectrum of non-viral transfection technologies for T cells. Strengths and weaknesses of each technology will be discussed in relation to T cell engineering. Altogether, this work emphasizes the potential of non-viral transfection approaches to advance the next-generation of genetically engineered T cells.
Collapse
Affiliation(s)
- Laurens Raes
- Laboratory of General Biochemistry & Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry & Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Koen Raemdonck
- Laboratory of General Biochemistry & Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Kevin Braeckmans
- Laboratory of General Biochemistry & Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| |
Collapse
|
6
|
CAR T cells: continuation in a revolution of immunotherapy. Lancet Oncol 2020; 21:e168-e178. [PMID: 32135120 DOI: 10.1016/s1470-2045(19)30823-x] [Citation(s) in RCA: 198] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/19/2019] [Accepted: 12/05/2019] [Indexed: 02/07/2023]
Abstract
The recent clinical successes of immunotherapy, as a result of a broader and more profound understanding of cancer immunobiology, and the leverage of this knowledge to effectively eradicate malignant cells, has revolutionised the field of cancer therapeutics. Immunotherapy is now considered the fifth pillar of cancer care, alongside surgery, chemotherapy, radiotherapy, and targeted therapy. Recently, the success of genetically modified T cells that express chimeric antigen receptors (CAR T cells) has generated considerable excitement. CAR T-cell therapy research and development has built on experience generated by laboratory research and clinical investigation of lymphokine-activated killer cells, tumour-infiltrating lymphocytes, and allogeneic haemopoietic stem-cell transplantation for cancer treatment. This Review aims to provide a background on the field of adoptive T-cell therapy and the development of genetically modified T cells, most notably CAR T-cell therapy. Many challenges exist to optimise efficacy, minimise toxicity, and broaden the application of immunotherapies based on T cells.
Collapse
|
7
|
Berg K, Schäfer VN, Bartnicki N, Eggenschwiler R, Cantz T, Stitz J. Rapid establishment of stable retroviral packaging cells and recombinant susceptible target cell lines employing novel transposon vectors derived from Sleeping Beauty. Virology 2019; 531:40-47. [DOI: 10.1016/j.virol.2019.02.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 02/06/2023]
|
8
|
Guirado E, Zhang Y, George A. Establishment of Stable Cell Lines from Primary Human Dental Pulp Stem Cells. Methods Mol Biol 2019; 1922:21-27. [PMID: 30838561 DOI: 10.1007/978-1-4939-9012-2_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
This protocol is for the isolation of primary human dental pulp stem cells (DPSCs) from adult extracted molars and for the generation of high-titer lentivirus for in vitro infection of the DPSCs. Stable cell lines of dental pulp stem cells are generated, maintained in culture, and used for subsequent experiments.
Collapse
Affiliation(s)
- Elizabeth Guirado
- Department of Oral Biology, College of Dentistry, The University of Illinois at Chicago, Chicago, IL, USA
| | - Youbin Zhang
- Department of Oral Biology, College of Dentistry, The University of Illinois at Chicago, Chicago, IL, USA
| | - Anne George
- Department of Oral Biology, College of Dentistry, The University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
9
|
Perica K, Curran KJ, Brentjens RJ, Giralt SA. Building a CAR Garage: Preparing for the Delivery of Commercial CAR T Cell Products at Memorial Sloan Kettering Cancer Center. Biol Blood Marrow Transplant 2018; 24:1135-1141. [PMID: 29499327 DOI: 10.1016/j.bbmt.2018.02.018] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 02/16/2018] [Indexed: 10/17/2022]
Abstract
Two commercial chimeric antigen receptor (CAR) T cell therapies for CD19-expressing B cell malignancies, Kymriah and Yescarta, have recently been approved by the Food and Drug Administration. The administration of CAR T cells is a complex endeavor involving cell manufacture, tracking and shipping of apheresis products, and management of novel and severe toxicities. At Memorial Sloan Kettering Cancer Center, we have identified 8 essential tasks that define the CAR T cell workflow. In this review, we discuss practical aspects of CAR T cell program development, including clinical, administrative, and regulatory challenges for successful implementation.
Collapse
Affiliation(s)
- Karlo Perica
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kevin J Curran
- Pediatric Bone Marrow Transplant Service, Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York; Cellular Therapeutics Center, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Renier J Brentjens
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Cellular Therapeutics Center, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sergio A Giralt
- Cellular Therapeutics Center, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, New York.
| |
Collapse
|
10
|
Flexible pseudotyping of retrovirus using recombinase-mediated cassette exchange. Biotechnol Lett 2018; 40:633-639. [PMID: 29353442 PMCID: PMC5862940 DOI: 10.1007/s10529-018-2515-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/11/2018] [Indexed: 10/27/2022]
Abstract
OBJECTIVE Develop an engineered cell line containing two flexible gene expression systems enabling the continuous production of tailor-made recombinant gammaretrovirus with predictable productivities through targeted integration. RESULTS Dual-FLEX cells (dFLEX) contain two independent recombinase-mediated cassette exchange (RMCE) systems which confer flexibility to the expression of different transgene and envelope combinations. The flexible envelope expression in dFLEX cells was validated by pseudotyping retrovirus particles with three different viral envelope proteins-GaLV, 4070A and VSV-G. Our results show that dFLEX cells are able to provide high titers of infectious retroviral particles with a single-copy integration of the envelope constructs after RMCE. The integrated CRE/Lox tagging cassette was amenable to express envelope proteins both using constitutive (i.e. CMV) and inducible (i.e. Tet-on) promoters. CONCLUSIONS dFLEX cell line provides predictable productivities of recombinant retrovirus pseudotyped with different envelope proteins broadening the tropism of particles that can be generated and thus accelerating the research and development of retrovirus-based products.
Collapse
|
11
|
Sadelain M, Rivière I, Riddell S. Therapeutic T cell engineering. Nature 2017; 545:423-431. [PMID: 28541315 DOI: 10.1038/nature22395] [Citation(s) in RCA: 583] [Impact Index Per Article: 83.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 04/26/2017] [Indexed: 12/13/2022]
Abstract
Genetically engineered T cells are powerful new medicines, offering hope for curative responses in patients with cancer. Chimaeric antigen receptors (CARs) are a class of synthetic receptors that reprogram lymphocyte specificity and function. CARs targeting CD19 have demonstrated remarkable potency in B cell malignancies. Engineered T cells are applicable in principle to many cancers, pending further progress to identify suitable target antigens, overcome immunosuppressive tumour microenvironments, reduce toxicities, and prevent antigen escape. Advances in the selection of optimal T cells, genetic engineering, and cell manufacturing are poised to broaden T-cell-based therapies and foster new applications in infectious diseases and autoimmunity.
Collapse
Affiliation(s)
- Michel Sadelain
- Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Isabelle Rivière
- Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Stanley Riddell
- Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| |
Collapse
|
12
|
Adams RM, Wang M, Crane AM, Brown B, Darlington GJ, Ledley FD. Effective Cryopreservation and Long-Term Storage of Primary Human Hepatocytes with Recovery of Viability, Differentiation, and Replicative Potential. Cell Transplant 2017; 4:579-86. [PMID: 8714779 DOI: 10.1177/096368979500400607] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Despite reports of successful cryopreservation of primary human hepatocytes, existing methods do not produce sufficient recovery of viable cells to meet the needs of basic research or clinical trials of hepatocellular transplantation. We now describe a protocol for efficient cryopreservation of primary human hepatocytes using University of Wisconsin (UW) solution, fetal bovine serum, and dimethyl sulfoxide (DMSO). This method provides >90% viability of differentiated, primary human hepatocytes 8 mo after cryopreservation as measured by trypan blue exclusion, preserves hepatocyte morphology, liver-specific gene expression α1 antitrypsin), and replication. The effectiveness of UW solution as a cryopreservative agent suggests that metabolic as well as ultrastructural factors may be important in the effective cryopreservation of primary human hepatocytes. The present method represents an effective protocol for cryopreserving differentiated primary human hepatocytes for research. This method may allow characterization and banking of human hepatocytes for clinical applications, including hepatocellular transplantation and hepatic assist devices.
Collapse
Affiliation(s)
- R M Adams
- Department of Cell Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|
13
|
Smith JA, Goldspiel BR. Cancer gene therapy update. J Oncol Pharm Pract 2016. [DOI: 10.1177/107815529900500101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective. To provide an update about gene marking and gene therapy trials in cancer patients. Data Sources. A MEDLINE search using the term “gene therapy” was conducted for the period 1985 to 1998. The reference lists from retrieved articles were reviewed. Meeting abstracts from the American Society of Clinical Oncology annual meeting (published in their proceedings) and the Annual Cancer Gene Therapy Symposium (published in Cancer Gene Therapy) that concerned gene therapy in cancer patients were also included. Data Extraction. Both authors reviewed the retrieved material and included preclinical data, case reports, and clinical trials related to gene transfer or gene therapy in cancer patients. Data Synthesis. There are several possible approaches to using gene therapy for the diagnosis and treatment of cancer and for the monitoring of cancer therapy. Exogenous genes may be used to mark cells to help better understand cancer biology or may be used directly for cancer treatment. Gene-marking trials have already provided new information about cancer biology and have demonstrated that reinfused progenitor cells may be a source of relapse in patients with acute or chronic myelogenous leukemia and neuroblastoma. Approaches using gene therapy for cancer treatment include: using lymphocytes as gene carriers, using foreign genes to increase tumor immunogenicity, introducing tumor regression antigen genes into viruses, introducing “sensitivity” genes to produce new cytotoxic agent(s) within tumors, producing new protein product(s) to protect normal cells, replacing missing or mutant tumor suppressor genes, and inactivating oncogenes. Clinical trials using these strategies have demonstrated that gene transfer is feasible (albeit with low transduction efficiency) and that gene expression occurs; in addition, clinical responses have been noted.
Collapse
Affiliation(s)
- Judith A Smith
- National Institutes of Health Clinical Center Pharmacy Department, Bethesda, Maryland
| | - Barry R Goldspiel
- National Institutes of Health Clinical Center Pharmacy Department, Bethesda, Maryland
| |
Collapse
|
14
|
|
15
|
Generation of a stable packaging cell line producing high-titer PPT-deleted integration-deficient lentiviral vectors. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2015; 2:15025. [PMID: 26229972 PMCID: PMC4510976 DOI: 10.1038/mtm.2015.25] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 06/08/2015] [Accepted: 06/11/2015] [Indexed: 12/29/2022]
Abstract
The risk of insertional mutagenesis inherent to all integrating exogenous expression cassettes was the impetus for the development of various integration-defective lentiviral vector (IDLV) systems. These systems were successfully employed in a plethora of preclinical applications, underscoring their clinical potential. However, current production of IDLVs by transient plasmid transfection is not optimal for large-scale production of clinical grade vectors. Here, we describe the development of the first tetracycline-inducible stable IDLV packaging cell line comprising the D64E integrase mutant and the VSV-G envelope protein. A conditional self-inactivating (cSIN) vector and a novel polypurine tract (PPT)-deleted vector were incorporated into the newly developed stable packaging cell line by transduction and stable transfection, respectively. High-titer (~10(7) infectious units (IU)/ml) cSIN vectors were routinely generated. Furthermore, screening of single-cell clones stably transfected with PPT-deleted vector DNA resulted in the identification of highly efficient producer cell lines generating IDLV titers higher than 10(8) IU/mL, which upon concentration increased to 10(10) IU/ml. IDLVs generated by stable producer lines efficiently transduce CNS tissues of rodents. Overall, the availability of high-titer IDLV lentivirus packaging cell line described here will significantly facilitate IDLV-based basic science research, as well as preclinical and clinical applications.
Collapse
|
16
|
Doi K, Takeuchi Y. [Gene therapy using retrovirus vectors: vector development and biosafety at clinical trials]. Uirusu 2015; 65:27-36. [PMID: 26923955 DOI: 10.2222/jsv.65.27] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Retrovirus vectors (gammaretroviral and lentiviral vectors) have been considered as promising tools to transfer therapeutic genes into patient cells because they can permanently integrate into host cellular genome. To treat monogenic, inherited diseases, retroviral vectors have been used to add correct genes into patient cells. Conventional gammaretroviral vectors achieved successful results in clinical trials: treated patients had therapeutic gene expression in target cells and had improved symptoms of diseases. However, serious side-effects of leukemia occurred, caused by retroviral insertional mutagenesis (IM). These incidences stressed the importance of monitoring vector integration sites in patient cells as well as of re-consideration on safer vectors. More recently lentiviral vectors which can deliver genes into non-dividing cells started to be used in clinical trials including neurological disorders, showing their efficacy. Vector integration site analysis revealed that lentiviruses integrate less likely to near promoter regions of oncogenes than gammaretroviruses and no adverse events have been reported in lentiviral vector-mediated gene therapy clinical trials. Therefore lentiviral vectors have promises to be applied to a wide range of common diseases in near future. For example, T cells from cancer patients were transduced to express chimeric T cell receptors recognizing their tumour cells enhancing patients' anti-cancer immunity.
Collapse
Affiliation(s)
- Knayo Doi
- MRC/UCL Centre for Medical Molecular Virology and Wohl Virion Centre, Division of infection and Immunity, University College London
| | | |
Collapse
|
17
|
Evans LH, Boi S, Malik F, Wehrly K, Peterson KE, Chesebro B. Analysis of two monoclonal antibodies reactive with envelope proteins of murine retroviruses: one pan specific antibody and one specific for Moloney leukemia virus. J Virol Methods 2014; 200:47-53. [PMID: 24556162 DOI: 10.1016/j.jviromet.2014.02.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 01/25/2014] [Accepted: 02/04/2014] [Indexed: 01/08/2023]
Abstract
Many monoclonal antibodies (MAbs) reactive with various proteins of murine leukemia viruses (MuLVs) have been developed. In this report two additional MAbs with differing and unusual specificities are described. MAb 573 is reactive with the envelope protein of all MuLVs tested including viruses in the ecotropic, xenotropic, polytropic and amphotropic classes. Notably, MAb 573 is one of only two reported MAbs that react with the envelope protein of amphotropic MuLVs. This MAb appears to recognize a conformational epitope within the envelope protein, as it reacts strongly with live virus and live infected cells, but does not react with formalin-fixed or alcohol-fixed infected cells or denatured viral envelope protein in immunoblots. In contrast, Mab 538 reacts only with an epitope unique to the envelope protein of the Moloney (Mo-) strain of MuLV, a prototypic ecotropic MuLV that is the basis for many retroviral tools used in molecular biology. MAb 538 can react with live cells and viruses, or detergent denatured or fixed envelope protein. The derivation of these antibodies as well as their characterization with regard to their isotype, range of reactivity with different MuLVs and utility in different immunological procedures are described in this study.
Collapse
Affiliation(s)
- Leonard H Evans
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT 59840, United States.
| | - Stefano Boi
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT 59840, United States; Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato (CA), Italy
| | - Frank Malik
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT 59840, United States
| | - Kathy Wehrly
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT 59840, United States
| | - Karin E Peterson
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT 59840, United States
| | - Bruce Chesebro
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT 59840, United States
| |
Collapse
|
18
|
Abstract
In this unit, the basic protocol generates stable cell lines that produce retroviral vectors that carry selectable markers. Also included are an alternate protocol that applies when the retroviral vector does not carry a selectable marker, and another alternate protocol for rapidly generating retroviral vector preparations by transient transfection. A support protocol describes construction of the retroviral vectors. The methods for generating virus from retroviral vector plasmids rely on the use of packaging cells that synthesize all of the retroviral proteins but do not produce replication-competent virus. Additional protocols detail plasmid transfection, virus titration, assay for replication-competent virus, and histochemical staining to detect transfer of a vector encoding alkaline phosphatase.
Collapse
Affiliation(s)
- A Dusty Miller
- Fred Hutchinson Cancer Research Center and Department of Pathology, University of Washington, Seattle, Washington
| |
Collapse
|
19
|
Schambach A, Zychlinski D, Ehrnstroem B, Baum C. Biosafety features of lentiviral vectors. Hum Gene Ther 2013; 24:132-42. [PMID: 23311447 DOI: 10.1089/hum.2012.229] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Over the past decades, lentiviral vectors have evolved as a benchmark tool for stable gene transfer into cells with a high replicative potential. Their relatively flexible genome and ability to transduce many forms of nondividing cells, combined with the potential for cell-specific pseudotyping, provides a rich resource for numerous applications in experimental platforms and therapeutic settings. Here, we give an overview of important biosafety features of lentiviral vectors, with detailed discussion of (i) the principles of the lentiviral split-genome design used for the construction of packaging cells; (ii) the relevance of modifications introduced into the lentiviral long terminal repeat (deletion of enhancer/promoter sequences and introduction of insulators); (iii) the basic features of mRNA processing, including the Rev/Rev-responsive element (RRE) interaction and the modifications of the 3' untranslated region of lentiviral vectors with various post-transcriptional regulatory elements affecting transcriptional termination, polyadenylation, and differentiation-specific degradation of mRNA; and (iv) the characteristic integration pattern with the associated risk of transcriptional interference with cellular genes. We conclude with considerations regarding the importance of cell targeting via envelope modifications. Along this course, we address canonical biosafety issues encountered with any type of viral vector: the risks of shedding, mobilization, germline transmission, immunogenicity, and insertional mutagenesis.
Collapse
Affiliation(s)
- Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, D-30625 Hannover, Germany
| | | | | | | |
Collapse
|
20
|
Garlick JA, Elias J, Taichman LB. Histochemical Detection of a Gene Transferred by Retrovirus Vector in Cultured Human Keratinocytes. J Histotechnol 2013. [DOI: 10.1179/his.1992.15.4.289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
21
|
Abstract
An understanding in the life cycle of γ-retroviruses has led to significant progress in the development of murine leukemia virus (MLV)-based vectors for gene delivery and human gene therapy. An MLV-based vector consists of the cis-acting sequences important for viral replication and gene expression. The sequence that encodes viral proteins is replaced with the gene of interest. To generate infectious retroviral vectors, viral-encoded proteins are supplied in trans for virion assembly. Here, we describe a method to rapidly generate MLV vectors from transiently transfected human 293T cells. The strategies to purify and titer the vector and to detect the presence of replication competent retrovirus (RCR) in the vector harvest are also described.
Collapse
Affiliation(s)
- Tammy Chang
- Department of Virology, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | | |
Collapse
|
22
|
Giacca M, Zacchigna S. Virus-mediated gene delivery for human gene therapy. J Control Release 2012; 161:377-88. [PMID: 22516095 DOI: 10.1016/j.jconrel.2012.04.008] [Citation(s) in RCA: 202] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Revised: 03/28/2012] [Accepted: 04/03/2012] [Indexed: 01/21/2023]
Abstract
After over 20 years from the first application of gene transfer in humans, gene therapy is now a mature discipline, which has progressively overcome several of the hurdles that prevented clinical success in the early stages of application. So far, the vast majority of gene therapy clinical trials have exploited viral vectors as very efficient nucleic acid delivery vehicles both in vivo and ex vivo. Here we summarize the current status of viral gene transfer for clinical applications, with special emphasis on the molecular properties of the major classes of viral vectors and the information so far obtained from gene therapy clinical trials.
Collapse
Affiliation(s)
- Mauro Giacca
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy.
| | | |
Collapse
|
23
|
Zakas PM, Spencer HT, Doering CB. Engineered Hematopoietic Stem Cells as Therapeutics for Hemophilia A. ACTA ACUST UNITED AC 2012; 1. [PMID: 25383239 DOI: 10.4172/2157-7412.s1-003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Philip M Zakas
- Graduate Program in Molecular and Systems Pharmacology, Graduate Division of Biological and Biomedical Sciences, Emory University
| | - H Trent Spencer
- Aflac Cancer Center and Blood Disorders Service, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Christopher B Doering
- Aflac Cancer Center and Blood Disorders Service, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
24
|
Liu XH, Xu W, Russ J, Eiden LE, Eiden MV. The host range of gammaretroviruses and gammaretroviral vectors includes post-mitotic neural cells. PLoS One 2011; 6:e18072. [PMID: 21464894 PMCID: PMC3065480 DOI: 10.1371/journal.pone.0018072] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Accepted: 02/20/2011] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Gammaretroviruses and gammaretroviral vectors, in contrast to lentiviruses and lentiviral vectors, are reported to be restricted in their ability to infect growth-arrested cells. The block to this restriction has never been clearly defined. The original assessment of the inability of gammaretroviruses and gammaretroviral vectors to infect growth-arrested cells was carried out using established cell lines that had been growth-arrested by chemical means, and has been generalized to neurons, which are post-mitotic. We re-examined the capability of gammaretroviruses and their derived vectors to efficiently infect terminally differentiated neuroendocrine cells and primary cortical neurons, a target of both experimental and therapeutic interest. METHODOLOGY/PRINCIPAL FINDINGS Using GFP expression as a marker for infection, we determined that both growth-arrested (NGF-differentiated) rat pheochromocytoma cells (PC12 cells) and primary rat cortical neurons could be efficiently transduced, and maintained long-term protein expression, after exposure to murine leukemia virus (MLV) and MLV-based retroviral vectors. Terminally differentiated PC12 cells transduced with a gammaretroviral vector encoding the anti-apoptotic protein Bcl-xL were protected from cell death induced by withdrawal of nerve growth factor (NGF), demonstrating gammaretroviral vector-mediated delivery and expression of genes at levels sufficient for therapeutic effect in non-dividing cells. Post-mitotic rat cortical neurons were also shown to be susceptible to transduction by murine replication-competent gammaretroviruses and gammaretroviral vectors. CONCLUSIONS/SIGNIFICANCE These findings suggest that the host range of gammaretroviruses includes post-mitotic and other growth-arrested cells in mammals, and have implications for re-direction of gammaretroviral gene therapy to neurological disease.
Collapse
Affiliation(s)
- Xiu-Huai Liu
- Section on Molecular Neuroscience, Laboratory of Cellular and Molecular Regulation, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Wenqin Xu
- Section on Directed Gene Transfer, Laboratory of Cellular and Molecular Regulation, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jill Russ
- Section on Directed Gene Transfer, Laboratory of Cellular and Molecular Regulation, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lee E. Eiden
- Section on Molecular Neuroscience, Laboratory of Cellular and Molecular Regulation, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Maribeth V. Eiden
- Section on Directed Gene Transfer, Laboratory of Cellular and Molecular Regulation, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
25
|
Liu J, Sumer H, Leung J, Upton K, Dottori M, Pébay A, Verma PJ. Late Passage Human Fibroblasts Induced to Pluripotency Are Capable of Directed Neuronal Differentiation. Cell Transplant 2011; 20:193-203. [DOI: 10.3727/096368910x514305] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
It is possible to generate induced pluripotent stem (iPS) cells from mouse and human somatic cells by ectopic expression of defined sets of transcription factors. However, the recommendation that somatic cells should be utilized at early passages for induced reprogramming limits their therapeutic application. Here we report successful reprogramming of human fibroblasts after more than 20 passages in vitro, to a pluripotent state with four transcription factors: Oct4, Sox2, Klf4, and c-Myc. The late passage-derived human iPS cells resemble human embryonic stem cells in morphology, cell surface antigens, pluripotent gene expression profiles, and epigenetic states. Moreover, these iPS cells differentiate into cell types representative of the three germ layers in teratomas in vivo, and directed neuronal differentiation in vitro.
Collapse
Affiliation(s)
- Jun Liu
- Centre for Reproduction and Development, Monash Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | - Huseyin Sumer
- Centre for Reproduction and Development, Monash Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | - Jessie Leung
- Centre for Neuroscience, The University of Melbourne, Parkville, Victoria, Australia
| | - Kyle Upton
- Centre for Reproduction and Development, Monash Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | - Mirella Dottori
- Centre for Neuroscience, The University of Melbourne, Parkville, Victoria, Australia
- Department of Pharmacology, The University of Melbourne, Parkville, Victoria, Australia
| | - Alice Pébay
- Centre for Neuroscience, The University of Melbourne, Parkville, Victoria, Australia
- Department of Pharmacology, The University of Melbourne, Parkville, Victoria, Australia
| | - Paul J. Verma
- Centre for Reproduction and Development, Monash Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
26
|
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
|
27
|
Doering CB, Archer D, Spencer HT. Delivery of nucleic acid therapeutics by genetically engineered hematopoietic stem cells. Adv Drug Deliv Rev 2010; 62:1204-12. [PMID: 20869414 PMCID: PMC2991563 DOI: 10.1016/j.addr.2010.09.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Revised: 08/17/2010] [Accepted: 09/08/2010] [Indexed: 01/02/2023]
Abstract
Several populations of adult human stem cells have been identified, but only a few of these are in routine clinical use. The hematopoietic stem cell (HSC) is arguably the most well characterized and the most routinely transplanted adult stem cell. Although details regarding several aspects of this cell's phenotype are not well understood, transplant of HSCs has advanced to become the standard of care for the treatment of a range of monogenic diseases and several types of cancer. It has also proven to be an excellent target for genetic manipulation, and clinical trials have already demonstrated the usefulness of targeting this cell as a means of delivering nucleic acid therapeutics for the treatment of several previously incurable diseases. It is anticipated that additional clinical trials will soon follow, such as genetically engineering HSCs with vectors to treat monogenic diseases such as hemophilia A. In addition to the direct targeting of HSCs, induced pluripotent stem (iPS) cells have the potential to replace virtually any engineered stem cell therapeutic, including HSCs. We now know that for the broad use of genetically modified HSCs for the treatment of non-lethal diseases, e.g. hemophilia A, we must be able to regulate the introduction of nucleic acid sequences into these target cells. We can begin to refine transduction protocols to provide safer approaches to genetically manipulate HSCs and strategies are being developed to improve the overall safety of gene transfer. This review focuses on recent advances in the systemic delivery of nucleic acid therapeutics using genetically modified stem cells, specifically focusing on i) the use of retroviral vectors to genetically modify HSCs, ii) the expression of fVIII from hematopoietic stem cells for the treatment of hemophilia A, and iii) the use of genetically engineered hematopoietic cells generated from iPS cells as treatment for disorders of hematopoiesis.
Collapse
Affiliation(s)
- Christopher B Doering
- Aflac Cancer Center and Blood Disorders Service, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | | | | |
Collapse
|
28
|
Abstract
Lentiviral vectors have become an important research tool and have just entered into clinical trials. As wild-type lentiviruses engage specific receptors that have limited tropism, most investigators have replaced the endogenous envelope glycoprotein with an alternative envelope. Such pseudotyped vectors have the potential to infect a wide variety of cell types and species. Alternatively, selection of certain viral envelope glycoproteins may also facilitate cell targeting to enhance directed gene transfer. We describe the method for generating pseudotyped vector and provide information regarding available pseudotypes and their respective target tissues.
Collapse
Affiliation(s)
- Daniela Bischof
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | |
Collapse
|
29
|
Koentgen F, Suess G, Naf D. Engineering the mouse genome to model human disease for drug discovery. Methods Mol Biol 2010; 602:55-77. [PMID: 20012392 DOI: 10.1007/978-1-60761-058-8_4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Genetically engineered mice (GEM) have become invaluable tools for human disease modeling and drug development. Completion of the mouse genome sequence in combination with transgenesis and gene targeting in embryonal stem cells have opened up unprecedented opportunities. Advanced technologies for derivation of GEM models will be introduced and discussed.
Collapse
|
30
|
Abstract
Retroviral vectors based on murine leukemia viruses (MuLV) have been used in clinical investigations for over a decade. Alternative retroviruses, most notably vectors based on HIV-1 and other lentiviruses, are now entering into clinical trials. Although vectors are designed to be replication defective, recombination events during vector production could lead to the generation of replication competent retroviruses (RCR) or replication competent lentiviruses (RCL). Careful screening of vector prior to human use must insure that patients are not inadvertently exposed to RCR or RCL. We describe methods capable of detecting low levels of virus contamination and discuss the current regulatory guidelines for screening gene therapy products intended for human use.
Collapse
Affiliation(s)
- Lakshmi Sastry
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | |
Collapse
|
31
|
McCarthy HO, Coulter JA, Robson T, Hirst DG. Gene therapy via inducible nitric oxide synthase: a tool for the treatment of a diverse range of pathological conditions. J Pharm Pharmacol 2008; 60:999-1017. [PMID: 18644193 DOI: 10.1211/jpp.60.8.0007] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nitric oxide (NO(.)) is a reactive nitrogen radical produced by the NO synthase (NOS) enzymes; it affects a plethora of downstream physiological and pathological processes. The past two decades have seen an explosion in the understanding of the role of NO(.) biology, highlighting various protective and damaging modes of action. Much of the controversy surrounding the role of NO(.) relates to the differing concentrations generated by the three isoforms of NOS. Both calcium-dependent isoforms of the enzyme (endothelial and neuronal NOS) generate low-nanomolar/picomolar concentrations of NO(.). By contrast, the calcium-independent isoform (inducible NOS (iNOS)) generates high concentrations of NO(.), 2-3 orders of magnitude greater. This review summarizes the current literature in relation to iNOS gene therapy for the therapeutic benefit of various pathological conditions, including various states of vascular disease, wound healing, erectile dysfunction, renal dysfunction and oncology. The available data provide convincing evidence that manipulation of endogenous NO(.) using iNOS gene therapy can provide the basis for future clinical trials.
Collapse
Affiliation(s)
- Helen O McCarthy
- School of Pharmacy, McClay Research Centre, Queen's University, Lisburn Road, Belfast, Northern Ireland, BT9 7BL, UK.
| | | | | | | |
Collapse
|
32
|
|
33
|
|
34
|
Abstract
In this unit, the basic protocol generates stable cell lines, which produce retroviral vectors that carry selectable markers. Alternate Protocol 1 applies when the retroviral vector does not carry a selectable marker. Alternate Protocol 2 presents rapidly generating retroviral vector preparations by transient transfection. Support Protocol 1 describes construction of the retrovirus vectors. The methods for generating virus from retroviral vector plasmids rely on the use of packaging cells that synthesize all of the retroviral proteins but do not produce replication-competent virus. One packaging cell line is transiently transfected to produce virus carrying the vector RNA that can be used to infect a second packaging cell line. Virus that carries a selectable marker can be titered and viral stocks can be tested for the presence of helper virus, as described. Vectors that contain sequence for alkaline phosphatase can be assayed histochemically. In this unit, the basic protocol generates stable cell lines, which produce retroviral vectors that carry selectable markers.
Collapse
Affiliation(s)
- A D Miller
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| |
Collapse
|
35
|
|
36
|
Hodgson CP, Chakraborty AK, Boman BM. Overview: Retroviral Vectors for Gene Therapy and Transgenics. ACTA ACUST UNITED AC 2008. [DOI: 10.1517/13543776.3.2.223] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
37
|
Haviernik P, Zhang Y, Bunting KD. Retroviral transduction of murine hematopoietic stem cells. Methods Mol Biol 2008; 430:229-41. [PMID: 18370303 DOI: 10.1007/978-1-59745-182-6_16] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Hematopoietic stem cells (HSC) are inherently rare cell types that cannot be obtained in sufficient amounts for classical biochemical characterization. To facilitate functional studies of murine HSC and hematopoietic development, the technique of retroviral-mediated gene transfer provides a useful tool. The generation of high titer retroviral vectors permits transduction of stem cells with a variety of genes and leads to long-term marking in the blood of recipient mice. Optimized promoter/enhancers facilitate high-level transgene expression in mice transplanted with transduced bone marrow (BM) cells. The co-expression of reporter genes along with a gene of interest greatly facilitates tracking donor engraftment of transduced hematopoietic progeny following stem cell transplantation. This methodology can be used to reconstitute defective function in a mutant background or to study protein function during hematopoiesis by overexpression. Despite limitations such as integration site variegation and copy number-dependent effects, this approach is rapid and efficient compared with transgenic mouse technology. In this chapter, we review this broadly applicable technique for achieving high-level murine BM stem cell transduction. We also describe methods for transplantation and subsequent analysis of transplanted mice as a bona fide assay for the stem cell transduction efficiency.
Collapse
Affiliation(s)
- Peter Haviernik
- Department of Medicine, Division of Hematology-Oncology, Center for Stem Cell and Regenerative Medicine, Case Western Reserve University, Cleveland, OH, USA
| | | | | |
Collapse
|
38
|
Tuszynski MH, Gage FH. Somatic gene therapy for nervous system disease. CIBA FOUNDATION SYMPOSIUM 2007; 196:85-94; discussion 94-7. [PMID: 8866129 DOI: 10.1002/9780470514863.ch7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Neurotrophic factors are target-derived molecules that prevent neuronal degeneration during development and, in some cases, during adulthood. They offer substantial promise as therapeutic agents in neurological disease by preventing cell loss and promoting axonal regeneration. However, the optimal means of delivering neurotrophic factors to the nervous system, and the CNS in particular, is an unresolved issue. Neurotrophic factors rarely influence only a single target neuronal population, hence broad delivery of neurotrophic factors to the nervous system may results in effects on multiple non-targeted neuronal populations. Ideally, neurotrophin delivery to the nervous system should be target-specific, regionally restricted, chronic, safe, well-tolerated and of sufficient concentration to elicit responses from target neurons. In this paper we discuss the use of somatic gene transfer methods to deliver neurotrophic factors to the CNS in a manner that seeks to meet the above criteria.
Collapse
Affiliation(s)
- M H Tuszynski
- Department of Neuroscience, University of California at San Diego, La Jolla 92093, USA
| | | |
Collapse
|
39
|
Dalba C, Bellier B, Kasahara N, Klatzmann D. Replication-competent Vectors and Empty Virus-like Particles: New Retroviral Vector Designs for Cancer Gene Therapy or Vaccines. Mol Ther 2007; 15:457-66. [PMID: 17245356 DOI: 10.1038/sj.mt.6300054] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Replication-defective vectors based on murine oncoretroviruses were the first gene transfer vectors to be used in successful gene therapies. Despite this achievement, they have two major drawbacks: insufficient efficacy for in vivo gene transfer and insertional mutagenesis. Attempts to overcome these problems have led to two retroviral vector designs of principally opposite character: replication-competent vectors transducing largely intact genomes and genome-free vectors. Replication-competent retroviral vectors have achieved dramatically improved efficacy for in vivo cancer gene therapy and genome-free retroviral vectors expressing different kinds of antigens have proven excellent as immunogens. Current developments aim to improve the safety of the replication-competent vectors and to augment the production efficiency of the genome-free vectors by expression from heterologous viral or non-viral vectors. Together with the continuous advances of classical defective retroviral vectors for ex vivo gene therapy, these developments illustrate that, due to their tremendous design versatility, retroviral vectors remain important vectors for gene therapy applications.
Collapse
Affiliation(s)
- Charlotte Dalba
- Biologie et Thérapeutique des Pathologies Immunitaires, CNRS, Université Pierre et Marie Curie-Paris, UMR 7087, Paris, France.
| | | | | | | |
Collapse
|
40
|
Reuss S, Biese P, Cosset FL, Takeuchi Y, Uckert W. Suspension packaging cell lines for the simplified generation of T-cell receptor encoding retrovirus vector particles. Gene Ther 2007; 14:595-603. [PMID: 17235289 DOI: 10.1038/sj.gt.3302906] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The transfer of T-cell receptor (TCR) genes into primary human T-cells to endow their specificity toward virus-infected and tumor cells is becoming an interesting tool for immunotherapy. TCR-modified T cells are mainly generated by retrovirus-mediated gene transfer. To produce TCR-retrovirus particles, fibroblast packaging cell lines are the most common tool. We constructed two packaging cell lines based on the human suspension T-cell lymphoma line Deltabeta-Jurkat, which lacks endogenous TCRbeta-chains and is therefore unable to express CD3 complexes on the cell surface. After supply of gag-pol (murine leukemia virus (Mo-MLV)) and env (GALV or MLV-10A1) genes, a green fluorescent protein (GFP)-encoding retrovirus vector was transduced into both packaging cell clones, which then stably produced GFP-retroviruses with titers of up to 4 x 10(5) infectious particles (IP)/ml. After transfer of a TCRalpha/beta-encoding retrovirus vector, Deltabeta-Jurkat/GALV and Deltabeta-Jurkat/10A1 cells expressed CD3 molecules on the cell surface. CD3-high expressing packaging cells were enriched by fluorescence-activated cell sorter sorting. In these cells, the CD3 expression level directly correlated with the titer of vector particles. TCR-retroviruses efficiently transduced human T-cell lines and primary T cells. In conclusion, the method allowed the fast and easy generation of high virus titer supernatants for TCR gene transfer.
Collapse
Affiliation(s)
- S Reuss
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | | | | | | | | |
Collapse
|
41
|
Baum C, Schambach A, Bohne J, Galla M. Retrovirus Vectors: Toward the Plentivirus? Mol Ther 2006; 13:1050-63. [PMID: 16632409 DOI: 10.1016/j.ymthe.2006.03.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2005] [Revised: 03/16/2006] [Accepted: 03/16/2006] [Indexed: 01/19/2023] Open
Abstract
Recombinant retroviral vectors based upon simple gammaretroviruses, complex lentiviruses, or potentially nonpathogenic spumaviruses represent relatively well characterized tools that are widely used for stable gene transfer. Different members of the Retroviridae family have developed distinct and potentially useful features related to their life cycle. These natural differences can be exploited for specialized applications in gene therapy and could conceivably be combined to create future retroviral hybrid vectors, ideally incorporating the following features: an efficient, noncytopathic packaging system with low likelihood of recombination; serum resistance; an ability to pseudotype with cell-specific envelopes; high-fidelity reverse transcription before cell entry; unrestricted cytoplasmic transport and nuclear import; an insulated expression cassette; specific chromosomal targeting; and physiologic or regulated levels of transgene expression. We envisage that, compared to contemporary vectors, a hybrid vector combining these properties would have increased therapeutic efficacy and an enhanced biosafety profile. Many of the above goals will require the inclusion of nonretroviral components into vector particles or transgenes.
Collapse
Affiliation(s)
- Christopher Baum
- Department of Experimental Hematology, Hannover Medical School, D-30625 Hannover, Germany.
| | | | | | | |
Collapse
|
42
|
|
43
|
Cornetta K, Matheson L, Ballas C. Retroviral vector production in the National Gene Vector Laboratory at Indiana University. Gene Ther 2006; 12 Suppl 1:S28-35. [PMID: 16231053 DOI: 10.1038/sj.gt.3302613] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The National Gene Vector Laboratory (NGVL) is a US National Institutes of Health initiative charged with providing clinical grade vectors for gene therapy trials. The program was started in 1995 and Indiana University has served as the production site for retroviral vectors and is also accepting applications for production of lentiviral vectors. The facility is designed to produce vectors for Phase I and Phase II clinical trials with the specific mandate to facilitate investigator-initiated research for academic institutions. To date, the facility has generated over 30 Master Cell Banks for gene therapy investigators throughout the United States. This required the facility to develop a system that can adapt to the varied needs of investigators, most of whom request different vector backbones, packaging cell lines, final product volumes, and media. In this review, we will illustrate some of the experiences of the Indiana University NGVL during the generation of retroviral vectors using murine-based packaging cell lines.
Collapse
Affiliation(s)
- K Cornetta
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | |
Collapse
|
44
|
Larochelle A, Krouse A, Metzger M, Orlic D, Donahue RE, Fricker S, Bridger G, Dunbar CE, Hematti P. AMD3100 mobilizes hematopoietic stem cells with long-term repopulating capacity in nonhuman primates. Blood 2006; 107:3772-8. [PMID: 16439684 PMCID: PMC1895780 DOI: 10.1182/blood-2005-09-3592] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
AMD3100, a bicyclam antagonist of the chemokine receptor CXCR4, has been shown to induce rapid mobilization of CD34(+) hematopoietic cells in mice, dogs, and humans, offering an alternative to G-CSF mobilization of peripheral-blood hematopoietic stem cells. In this study, AMD3100-mobilized CD34(+) cells were phenotypically analyzed, marked with Neo(R)-containing retroviral vectors, and subsequently transplanted into myeloablated rhesus macaques. We show engraftment of transduced AMD3100-mobilized CD34(+) cells with Neo(R) gene marked myeloid and lymphoid cells up to 32 months after transplantation, demonstrating the ability of AMD3100 to mobilize true long-term repopulating hematopoietic stem cells. More AMD3100-mobilized CD34(+) cells are in the G(1) phase of the cell cycle and more cells express CXCR4 and VLA-4 compared with G-CSF-mobilized CD34(+) cells. In vivo gene marking levels obtained with AMD3100-mobilized CD34(+) cells were better than those obtained using CD34(+) cells mobilized with G-CSF alone. Overall, these results indicate that AMD3100 mobilizes a population of hematopoietic stem cells with intrinsic characteristics different from those of hematopoietic stem cells mobilized with G-CSF, suggesting fundamental differences in the mechanism of AMD3100-mediated and G-CSF-mediated hematopoietic stem cell mobilization. Thus, AMD3100-mobilized CD34(+) cells represent an alternative source of hematopoietic stem cells for clinical stem cell transplantation and genetic manipulation with integrating retroviral vectors.
Collapse
Affiliation(s)
- André Larochelle
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bldg 10 CRC, Rm 4E-5132, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
El-Andaloussi S, Johansson H, Magnusdottir A, Järver P, Lundberg P, Langel U. TP10, a delivery vector for decoy oligonucleotides targeting the Myc protein. J Control Release 2005; 110:189-201. [PMID: 16253378 DOI: 10.1016/j.jconrel.2005.09.012] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2005] [Revised: 09/07/2005] [Accepted: 09/13/2005] [Indexed: 11/26/2022]
Abstract
One approach to investigate gene function, by silencing the activity of certain proteins, is the usage of double stranded decoy oligodeoxynucleotides (ds decoy ODNs). Decoy, in this sense, is ds ODNs bearing the consensus binding sequence for a DNA-binding protein. This can be used in clinical settings to attenuate the effect of overexpressed transcription factors in tumor cells. We here choose to target the oncogenic protein Myc. Since oligonucleotides are poorly internalized to cells, a cell-penetrating peptide, TP10, was coupled to the Myc decoy, using two different strategies. Either TP10 was simply mixed with ds decoy ODNs forming complexes through non-covalent electrostatic interactions, or by having a nona-nucleotide overhang in one of the decoy strands, and adding a complementary PNA sequence coupled to an NLS sequence and TP10, which could hybridize to the Myc decoy. By using these strategies, uptake was significantly enhanced, especially with the co-incubation approach. Interestingly, various endocytosis inhibitors had no effect on the uptake pattern, suggesting that uptake of these complexes is not mediated via endocytosis. Finally, a decreased proliferative capacity was observed when treating the neuroblastoma cell line N2a with TP10-PNA conjugate hybridized to Myc decoy compared to naked Myc decoy and untreated cells. A dose-dependent decrease in proliferation was also observed in MCF-7 cells, when using both strategies. These results suggest an alternative way to efficiently deliver ds ODNs into cells using the cell-penetrating peptide TP10 and prevent tumor growth by targeting the oncogenic protein Myc.
Collapse
Affiliation(s)
- S El-Andaloussi
- Department of Neurotoxicology, Stockholm University, Sweden.
| | | | | | | | | | | |
Collapse
|
46
|
|
47
|
Gonçalves MAFV. A concise peer into the background, initial thoughts and practices of human gene therapy. Bioessays 2005; 27:506-17. [PMID: 15832383 DOI: 10.1002/bies.20218] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The concept of human gene therapy came on the heels of fundamental discoveries on the nature and working of the gene. However, realistic prospects to correct the underlying cause of recessive genetic disorders through the transfer of wild-type alleles of defective genes had to wait for the arrival of recombinant DNA technology. These techniques permitted the isolation and insertion of genes into the first recombinant delivery systems. The realization that viruses are natural gene carriers provided inspiration for gene therapy and, as engineered vectors, viruses became prominent gene delivery vehicles. Nonetheless, when put in the context of human and non-human primate studies, all vectors fell short of success regardless of their viral or non-viral origin. Recognition of issues such as inefficient gene transfer and short-lived or scant expression in the relevant cell type(s) prompted researchers to refine and develop several gene delivery systems, in particular those based on retroviruses, adeno-associated viruses and adenoviruses. Concomitantly, available technology was deployed to tackle disorders that require few genetically corrected cells to attain therapy.
Collapse
Affiliation(s)
- Manuel A F V Gonçalves
- Gene Therapy Section, Department of Molecular Cell Biology, Leiden University Medical Center, Wassenaarseweg 72, 2333 AL Leiden, the Netherlands.
| |
Collapse
|
48
|
Fukuda S, Broxmeyer HE, Pelus LM. Flt3 ligand and the Flt3 receptor regulate hematopoietic cell migration by modulating the SDF-1α(CXCL12)/CXCR4 axis. Blood 2005; 105:3117-26. [PMID: 15618475 DOI: 10.1182/blood-2004-04-1440] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
AbstractFlt3 ligand (FL) enhances hematopoietic cell proliferation and facilitates hematopoietic stem cell mobilization in vivo, while the stromal-derived factor 1α (SDF-1α, CXC ligand 12 [CXCL12])/CXC receptor 4 (CXCR4) axis is critical for their homing and trafficking. We investigated if FL and its receptor, Flt3, functionally interact with CXCL12/CXCR4 to regulate hematopoietic cell migration. FL stimulated chemokinetic activity when used alone, but synergistically enhanced short-term migration of CD34+ cells, Ba/F3 cells expressing human Flt3 (Ba/F3-Flt3), and human RS4;11 acute leukemia cells, induced by CXCL12. Moreover, overexpression of constitutively activated internal tandem duplication (ITD)–Flt3 mutants in Ba/F3 cells dramatically enhanced migration toward CXCL12. In Ba/F3-Flt3 cells, synergistic cell migration to FL plus CXCL12 was associated with enhanced phosphorylation of mitogen-activated protein kinase p42/p44 (MAPKp42/p44), cyclic adenosine monophosphate response element binding protein (CREB), and Akt, and was partially inhibited by pretreatment of cells with selective inhibitors for MAPKp42/p44, protein kinase A (PKA), or phosphatidylinositol 3–kinase (PI3-kinase), implicating these pathways in migration to FL plus CXCL12. In contrast, prolonged exposure of CD34+ or Ba/F3-Flt3 cells to FL down-regulated CXCR4 expression, inhibited CXCL12-mediated phosphorylation of MAPKp42/p44, CREB, and Akt, and impaired migration toward CXCL12. These findings suggest that FL/Flt3 may facilitate hematopoietic cell migration/homing and mobilization by enhancing or inhibiting CXCL12/CXCR4 signaling pathways and that the FL/Flt3 axis participates in trafficking of normal and transformed hematopoietic cells.
Collapse
Affiliation(s)
- Seiji Fukuda
- Department of Microbiology & Immunology and the Walther Oncology Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | | | | |
Collapse
|
49
|
Targeted gene delivery: The role of peptide nucleic acid. Int J Pept Res Ther 2005. [DOI: 10.1007/s10989-005-4922-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
50
|
Hahm SH, Yi Y, Lee DK, Noh MJ, Yun L, Hwang S, Lee KH. Construction of retroviral vectors with enhanced efficiency of transgene expression. J Virol Methods 2004; 121:127-36. [PMID: 15381349 DOI: 10.1016/j.jviromet.2004.05.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2004] [Revised: 05/19/2004] [Accepted: 05/27/2004] [Indexed: 11/19/2022]
Abstract
Retroviral vectors have been widely used in gene therapy due to their simple genomic structure and high transduction efficiency. We report a construction of Moloney murine sarcoma virus (MoMSV) and Moloney murine leukemia virus (MoMLV) hybrid-based retroviral vectors with significantly improved efficiency of transgene expression after stable incorporation into the host genome. In these vectors, the residual gag gene coding sequence located in the extended region of packaging signal was removed. These vectors, therefore, contain no coding sequence for the gag, pol, or env gene that can be used for homologous recombination with sequences introduced in the packaging system for a recombinant competent retrovirus (RCR) generation. A strong splice acceptor site obtained from the exon/intron junction of either the chimpanzee EF1-alpha gene or the human CMV major immediate early gene was placed downstream of the MoMSV packaging signal (Psi), significantly improving the efficiency of transgene expression. The 5' LTR U3 sequence was replaced with an extended human CMV major immediate early gene enhancer/promoter for a strong expression of full-length messages from the viral backbone, helping to maintain high levels of viral titer. These newly developed retroviral vectors should facilitate RCR-free gene transfer with significantly improved efficacy in clinical gene therapy trials.
Collapse
|