1
|
Lotter C, Kuzucu EÜ, Casper J, Alter CL, Puligilla RD, Detampel P, Lopez JS, Ham AS, Huwyler J. Comparison of ionizable lipids for lipid nanoparticle mediated DNA delivery. Eur J Pharm Sci 2024; 203:106898. [PMID: 39260517 DOI: 10.1016/j.ejps.2024.106898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/26/2024] [Accepted: 09/09/2024] [Indexed: 09/13/2024]
Abstract
Lipid nanoparticles (LNPs) are successfully used for RNA-based gene delivery. In the context of gene replacement therapies, however, delivery of DNA expression plasmids using LNPs as a non-viral vector could be a promising strategy for the induction of longer-lasting effects. Therefore, DNA expression plasmids (3 to 4 kbp) coding for fluorescent markers or luciferase were combined with LNPs. Different clinically used ionizable lipids (DLin-MC3-DMA, SM-102, and ALC-0315) were tested to compare their influence on DNA plasmid delivery. DNA-LNPs were characterized with respect to their colloidal properties (size, polydispersity, ζ-potential, morphology), in vitro performance (cellular uptake, DNA delivery, and gene expression), and in vivo characteristics (biodistribution and luciferase gene expression). At an optimized N/P ratio of 6, spherical, small and monodisperse particles with anionic ζ-potential were obtained. Efficient transgene expression was achieved with a minimum amount of 1 pg DNA per initially plated cells. Zebrafish studies allowed selection of DNA-LNPs, which demonstrated prolonged blood circulation, avoidance of macrophage clearance, and vascular extravasation. Our comparative study demonstrates a high impact of the ionizable lipid type on DNA-LNP performance. Superior transfection efficiency of DNA-LNPs containing the ionizable lipid ALC-0315 was confirmed in wildtype mice.
Collapse
Affiliation(s)
- Claudia Lotter
- Pharmazentrum, Division of Pharmaceutical Technology, University of Basel, CH-4056 Basel, Switzerland
| | - Evrim Ümit Kuzucu
- Pharmazentrum, Division of Pharmaceutical Technology, University of Basel, CH-4056 Basel, Switzerland
| | - Jens Casper
- Pharmazentrum, Division of Pharmaceutical Technology, University of Basel, CH-4056 Basel, Switzerland
| | - Claudio Luca Alter
- Pharmazentrum, Division of Pharmaceutical Technology, University of Basel, CH-4056 Basel, Switzerland; Swiss Nanoscience Institute, University of Basel, CH-4056 Basel, Switzerland
| | - Ramya Deepthi Puligilla
- Pharmazentrum, Division of Pharmaceutical Technology, University of Basel, CH-4056 Basel, Switzerland
| | - Pascal Detampel
- Pharmazentrum, Division of Pharmaceutical Technology, University of Basel, CH-4056 Basel, Switzerland
| | - Juana Serrano Lopez
- Instituto Investigación Sanitaria Fundación Jiménez Díaz, ES-28015 Madrid, Spain
| | | | - Jörg Huwyler
- Pharmazentrum, Division of Pharmaceutical Technology, University of Basel, CH-4056 Basel, Switzerland.
| |
Collapse
|
2
|
Hsieh EWY, Bolze A, Hernandez JD. Inborn errors of immunity illuminate mechanisms of human immunology and pave the road to precision medicine. Immunol Rev 2024; 322:5-14. [PMID: 38308392 DOI: 10.1111/imr.13311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2024]
Affiliation(s)
- Elena W Y Hsieh
- Department of Pediatrics, Section of Allergy and Immunology, Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Children's Hospital Colorado, Aurora, Colorado, USA
| | | | - Joseph D Hernandez
- Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics, Stanford University, Stanford, California, USA
| |
Collapse
|
3
|
Ghanim HY, Porteus MH. Gene regulation in inborn errors of immunity: Implications for gene therapy design and efficacy. Immunol Rev 2024; 322:157-177. [PMID: 38233996 DOI: 10.1111/imr.13305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/28/2023] [Accepted: 01/02/2024] [Indexed: 01/19/2024]
Abstract
Inborn errors of immunity (IEI) present a unique paradigm in the realm of gene therapy, emphasizing the need for precision in therapeutic design. As gene therapy transitions from broad-spectrum gene addition to careful modification of specific genes, the enduring safety and effectiveness of these therapies in clinical settings have become crucial. This review discusses the significance of IEIs as foundational models for pioneering and refining precision medicine. We explore the capabilities of gene addition and gene correction platforms in modifying the DNA sequence of primary cells tailored for IEIs. The review uses four specific IEIs to highlight key issues in gene therapy strategies: X-linked agammaglobulinemia (XLA), X-linked chronic granulomatous disease (X-CGD), X-linked hyper IgM syndrome (XHIGM), and immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX). We detail the regulatory intricacies and therapeutic innovations for each disorder, incorporating insights from relevant clinical trials. For most IEIs, regulated expression is a vital aspect of the underlying biology, and we discuss the importance of endogenous regulation in developing gene therapy strategies.
Collapse
Affiliation(s)
- Hana Y Ghanim
- Division of Pediatrics, Division of Oncology, Hematology, Stem Cell Transplantation, Stanford University, Stanford, California, USA
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Matthew H Porteus
- Division of Pediatrics, Division of Oncology, Hematology, Stem Cell Transplantation, Stanford University, Stanford, California, USA
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
- Center for Definitive and Curative Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
4
|
Whitney D, Shestopalov I, Fincker M, d’Anjou M, Kral K, Gayron M, Pierciey FJ, Colvin RA. Drug product attributes predict clinical efficacy in betibeglogene autotemcel gene therapy for β-thalassemia. Mol Ther Methods Clin Dev 2023; 31:101155. [PMID: 38074412 PMCID: PMC10709156 DOI: 10.1016/j.omtm.2023.101155] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 11/08/2023] [Indexed: 10/16/2024]
Abstract
Ex vivo autologous hematopoietic stem cell lentiviral-based gene therapy with betibeglogene autotemcel has been studied in patients with transfusion-dependent β-thalassemia in Phase III clinical trials (HGB-207 and HGB-212), with 90% of patients reaching transfusion independence (TI). Here, we explore manufacturing parameters, drug product quality attributes, and limited patient characteristics that had an impact on clinical efficacy in HGB-207 and HGB-212. Retrospective analysis revealed that the peripheral blood vector copy number (VCN) was related to TI, with a strong correlation between peripheral blood VCN at 6 months and gene therapy-derived therapeutic protein (HbAT87Q) expression at 6 months (correlation coefficient, 0.8681; p < 0.0001; R2 = 0.7536). A peripheral blood VCN threshold of ≥0.75 copies per diploid genome at 6 months post betibeglogene autotemcel infusion provided a stringent surrogate biomarker for TI and was used as the outcome variable for multivariate analysis using a random forest classifier. The top predictive feature of clinical efficacy was found to be the percentage of lentiviral vector-positive cells in the drug product. This retrospective analysis is critical to understanding the key product quality attributes that can predict clinical efficacy in lentiviral vector gene therapy within this clinical trial population.
Collapse
Affiliation(s)
| | | | | | | | - Kelly Kral
- bluebird bio, Inc., Somerville, MA 02145, USA
| | | | | | | |
Collapse
|
5
|
Wang Y, Zhou SK, Wang Y, Lu ZD, Zhang Y, Xu CF, Wang J. Engineering tumor-specific gene nanomedicine to recruit and activate T cells for enhanced immunotherapy. Nat Commun 2023; 14:1993. [PMID: 37031188 PMCID: PMC10082825 DOI: 10.1038/s41467-023-37656-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 03/27/2023] [Indexed: 04/10/2023] Open
Abstract
PD-1/PD-L1 blockade therapy that eliminates T-cell inhibition signals is successful, but poor benefits are often observed. Increasing T-cell infiltration and quantity of PD-1/PD-L1 inhibitors in tumor can improve efficacy but remains challenging. Here, we devise tumor-specific gene nanomedicines to mobilize tumor cells to secrete CXCL9 (T-cell chemokine) and anti-PD-L1 scFv (αPD-L1, PD-L1 blocking agent) for enhanced immunotherapy. The tyrosinase promoter-driven NPTyr-C9AP can specifically co-express CXCL9 and αPD-L1 in melanoma cells, thereby forming a CXCL9 gradient for T-cell recruitment and high intratumoral αPD-L1 concentration for enhancing T-cell activation. As a result, NPTyr-C9AP shows strong antimelanoma effects. Moreover, specific co-expression of CXCL9 and αPD-L1 in various tumor cells is achieved by replacing the tyrosinase promoter of NPTyr-C9AP with a survivin promoter, which increases T-cell infiltration and activation and therapeutic efficacy in multiple tumors in female mice. This study provides a strategy to maximize the immunotherapeutic outcome regardless of the heterogeneous tumor microenvironment.
Collapse
Affiliation(s)
- Yue Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P.R. China
| | - Shi-Kun Zhou
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P.R. China
| | - Yan Wang
- School of Medicine, South China University of Technology, Guangzhou, 510006, P.R. China
| | - Zi-Dong Lu
- School of Medicine, South China University of Technology, Guangzhou, 510006, P.R. China
| | - Yue Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P.R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P.R. China
| | - Cong-Fei Xu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P.R. China.
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P.R. China.
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, P.R. China.
| | - Jun Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P.R. China.
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P.R. China.
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, P.R. China.
| |
Collapse
|
6
|
White M, Whittaker RG. Post-Trial Considerations for an Early Phase Optogenetic Trial in the Human Brain. OPEN ACCESS JOURNAL OF CLINICAL TRIALS 2022. [DOI: 10.2147/oajct.s345482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
7
|
Müller TR, Jarosch S, Hammel M, Leube J, Grassmann S, Bernard B, Effenberger M, Andrä I, Chaudhry MZ, Käuferle T, Malo A, Cicin-Sain L, Steinberger P, Feuchtinger T, Protzer U, Schumann K, Neuenhahn M, Schober K, Busch DH. Targeted T cell receptor gene editing provides predictable T cell product function for immunotherapy. Cell Rep Med 2021; 2:100374. [PMID: 34467251 PMCID: PMC8385324 DOI: 10.1016/j.xcrm.2021.100374] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 06/15/2021] [Accepted: 07/20/2021] [Indexed: 01/02/2023]
Abstract
Adoptive transfer of T cells expressing a transgenic T cell receptor (TCR) has the potential to revolutionize immunotherapy of infectious diseases and cancer. However, the generation of defined TCR-transgenic T cell medicinal products with predictable in vivo function still poses a major challenge and limits broader and more successful application of this "living drug." Here, by studying 51 different TCRs, we show that conventional genetic engineering by viral transduction leads to variable TCR expression and functionality as a result of variable transgene copy numbers and untargeted transgene integration. In contrast, CRISPR/Cas9-mediated TCR replacement enables defined, targeted TCR transgene insertion into the TCR gene locus. Thereby, T cell products display more homogeneous TCR expression similar to physiological T cells. Importantly, increased T cell product homogeneity after targeted TCR gene editing correlates with predictable in vivo T cell responses, which represents a crucial aspect for clinical application in adoptive T cell immunotherapy.
Collapse
Affiliation(s)
- Thomas R. Müller
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Sebastian Jarosch
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), Munich, Germany
| | - Monika Hammel
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), Munich, Germany
| | - Justin Leube
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), Munich, Germany
| | - Simon Grassmann
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), Munich, Germany
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Bettina Bernard
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), Munich, Germany
| | - Manuel Effenberger
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), Munich, Germany
| | - Immanuel Andrä
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), Munich, Germany
| | - M. Zeeshan Chaudhry
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, Braunschweig, Germany
| | - Theresa Käuferle
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
- Department of Pediatric Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, Germany
| | - Antje Malo
- Institute of Virology, TUM, Munich, Germany
| | - Luka Cicin-Sain
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, Braunschweig, Germany
| | - Peter Steinberger
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Tobias Feuchtinger
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
- Department of Pediatric Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, Germany
| | - Ulrike Protzer
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
- Institute of Virology, TUM, Munich, Germany
| | - Kathrin Schumann
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), Munich, Germany
- Institute for Advanced Study, TUM, Munich, Germany
| | - Michael Neuenhahn
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Kilian Schober
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), Munich, Germany
| | - Dirk H. Busch
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
- Institute for Advanced Study, TUM, Munich, Germany
| |
Collapse
|
8
|
Lyu Y, Yang C, Lyu X, Pu K. Active Delivery of CRISPR System Using Targetable or Controllable Nanocarriers. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2005222. [PMID: 33759340 DOI: 10.1002/smll.202005222] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/30/2020] [Indexed: 05/17/2023]
Abstract
Among programmable nuclease-based genome editing tools, the clustered regularly interspaced short palindromic repeats (CRISPR) system with accuracy and the convenient operation is most promising to be applied in gene therapy. The development of effective delivery carriers for the CRISPR system is the major premise to achieve practical applications. Although many nanocarrier-mediated deliveries have been reported to be safer and cheaper over the physical and viral delivery, the accumulation at disease sites or controllability with the spatial or temporal resolution are still desired on nanocarriers to reduce side effects and off-target from the CRISPR system. Therefore, the targetable and controllable nanocarriers to actively deliver the CRISPR system are summarized. The cell or even organ selective nanocarriers are introduced first, followed by the discussion of nanocarriers controlled by biochemical or physical signals. At last, the potential challenges faced by existing nanocarriers are discussed.
Collapse
Affiliation(s)
- Yan Lyu
- Cosmetic Innovation Center, School of Chemical and Material Engineering, Jiangnan University, Wuxi, 214122, China
- Key Laboratory of Synthetic and Biological Colloids Ministry of Education, School of Chemical and Material Engineering, Jiangnan University, Wuxi, 214122, China
| | - Cheng Yang
- Cosmetic Innovation Center, School of Chemical and Material Engineering, Jiangnan University, Wuxi, 214122, China
- Key Laboratory of Synthetic and Biological Colloids Ministry of Education, School of Chemical and Material Engineering, Jiangnan University, Wuxi, 214122, China
| | - Xiaomei Lyu
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Kanyi Pu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637457, Singapore
| |
Collapse
|
9
|
Chimeric Antigen Receptor T-cell Therapy for Multiple Myeloma. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 21:21-34. [PMID: 33046423 DOI: 10.1016/j.clml.2020.08.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/27/2020] [Accepted: 08/29/2020] [Indexed: 12/17/2022]
Abstract
Relapsed/refractory multiple myeloma (MM) remains a significant clinical challenge, despite a wide array of approved therapeutic agents. Immunotherapy offers an advantage in this setting. Chimeric antigen receptor (CAR) modified T-cells have transformed care for patients with hematologic malignancies. CAR-T cells targeting CD-19 B-cell lymphoma cells have shown prominent activity in lymphoma and acute lymphoblastic leukemia. Recently, the CAR-T cell platform for MM demonstrated therapeutic benefit. Hence, it is rapidly progressing. The most commonly tested target for MM is the B-cell maturation antigen. Complexities involved in the generation and use of CAR-T cells for MM include the identification of appropriate target antigens that are specific, and tumor type restricted, in addition to the optimization of CAR constructs to mitigate toxicities including cytokine release syndrome. CAR-T cells hold immense promise as a therapeutic modality for the treatment of MM. In this article, we provide an updated review of clinical trials of MM-specific CAR-T cells.
Collapse
|
10
|
Uddin F, Rudin CM, Sen T. CRISPR Gene Therapy: Applications, Limitations, and Implications for the Future. Front Oncol 2020; 10:1387. [PMID: 32850447 PMCID: PMC7427626 DOI: 10.3389/fonc.2020.01387] [Citation(s) in RCA: 224] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 06/30/2020] [Indexed: 12/24/2022] Open
Abstract
A series of recent discoveries harnessing the adaptive immune system of prokaryotes to perform targeted genome editing is having a transformative influence across the biological sciences. The discovery of Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and CRISPR-associated (Cas) proteins has expanded the applications of genetic research in thousands of laboratories across the globe and is redefining our approach to gene therapy. Traditional gene therapy has raised some concerns, as its reliance on viral vector delivery of therapeutic transgenes can cause both insertional oncogenesis and immunogenic toxicity. While viral vectors remain a key delivery vehicle, CRISPR technology provides a relatively simple and efficient alternative for site-specific gene editing, obliviating some concerns raised by traditional gene therapy. Although it has apparent advantages, CRISPR/Cas9 brings its own set of limitations which must be addressed for safe and efficient clinical translation. This review focuses on the evolution of gene therapy and the role of CRISPR in shifting the gene therapy paradigm. We review the emerging data of recent gene therapy trials and consider the best strategy to move forward with this powerful but still relatively new technology.
Collapse
Affiliation(s)
- Fathema Uddin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Charles M. Rudin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Weill Cornell Medicine, Cornell University, New York, NY, United States
| | - Triparna Sen
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Weill Cornell Medicine, Cornell University, New York, NY, United States
| |
Collapse
|
11
|
White M, Mackay M, Whittaker R. Taking Optogenetics into the Human Brain: Opportunities and Challenges in Clinical Trial Design. OPEN ACCESS JOURNAL OF CLINICAL TRIALS 2020; 2020:33-41. [PMID: 34471390 DOI: 10.2147/oajct.s259702] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Optogenetics, the use of light to control the activity of suitably sensitized cells, has led to major advances in the field of basic neuroscience since it first emerged in 2005. Already, the technique has entered clinical trials for conditions such as Retinitis Pigmentosa. A major focus of interest is the use of optogenetics within the brain, where the ability to precisely control the activity of specific subsets of neurons could lead to novel treatments for a wide range of disorders from epilepsy to schizophrenia. However, since any therapy would require both the use of gene therapy techniques to introduce non-human proteins, and implantable electronic devices to provide optical stimulation, applying this technique in the brain presents a unique set of obstacles and challenges. This review looks at the reasons why researchers are exploring the use of optogenetics within the brain. It then explores the challenges facing scientists, engineers and clinicians wanting to take this technology from the lab into the first human brain, discussing different possibilities for a first-in-human clinical trial from a sponsor, patient and regulatory perspective.
Collapse
Affiliation(s)
- Michael White
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, United Kingdom
| | - Michael Mackay
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, United Kingdom
| | - Roger Whittaker
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, United Kingdom.,Department of Clinical Neurophysiology, Royal Victoria Hospital, Queen Victoria Road, Newcastle upon Tyne, NE1 4LP, United Kingdom
| |
Collapse
|
12
|
Chiang CY, Ligunas GD, Chin WC, Ni CW. Efficient Nonviral Stable Transgenesis Mediated by Retroviral Integrase. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 17:1061-1070. [PMID: 32462054 PMCID: PMC7240061 DOI: 10.1016/j.omtm.2020.04.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 04/27/2020] [Indexed: 11/28/2022]
Abstract
Efficient transgene delivery is critical for genetic manipulation and therapeutic intervention of target cells. Two well-characterized integrative systems have been described that rely on viral and nonviral vectors. However, use of viral vectors for gene therapy has been associated with several safety concerns. Here, we report a virus-free method for stable transgenesis based on the reaction of retroviral integrase. We constructed a gateway cloning compatible vector containing two truncated long terminal repeat (LTR) sequences (dLTR) that flank the transgene cassette. Notably, 5′-ACTG-3′ and blunt-end restriction cutting sites were also embedded at the end of dLTR to be recognized by HIV-1 integrase. When performing coinjection of transgene cassette and integrase mRNA into zebrafish embryos at one cell stage, there were 50% to 55% of injected embryos expressing a marker gene in a desired pattern. When applying our method in mammalian cells, there were 42% of cultured human epithelial cell lines showing stable integration. These results demonstrated that our method can successfully insert an exogenous gene into the host genome with highly efficient integration. Importantly, this system operates without most of the viral components while retaining effective stable transgenesis. We anticipate this method will provide a convenient, safe, and highly efficient way for applications in transgenesis and gene therapy.
Collapse
Affiliation(s)
- Chang-Ying Chiang
- Department of Bioengineering, School of Engineering, University of California, Merced, Merced, CA, USA
| | - Gloria Denise Ligunas
- Program of Quantitative and Systems Biology, University of California, Merced, Merced, CA, USA
| | - Wei-Chun Chin
- Department of Bioengineering, School of Engineering, University of California, Merced, Merced, CA, USA.,Program of Quantitative and Systems Biology, University of California, Merced, Merced, CA, USA
| | - Chih-Wen Ni
- Department of Bioengineering, School of Engineering, University of California, Merced, Merced, CA, USA.,Program of Quantitative and Systems Biology, University of California, Merced, Merced, CA, USA
| |
Collapse
|
13
|
Santeramo I, Bagnati M, Harvey EJ, Hassan E, Surmacz-Cordle B, Marshall D, Di Cerbo V. Vector Copy Distribution at a Single-Cell Level Enhances Analytical Characterization of Gene-Modified Cell Therapies. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 17:944-956. [PMID: 32420408 PMCID: PMC7217927 DOI: 10.1016/j.omtm.2020.04.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 04/22/2020] [Indexed: 12/28/2022]
Abstract
The ability to deliver transgenes into the human genome using viral vectors is a major enabler of the gene-modified cell therapy field. However, the control of viral transduction is difficult and can lead to product heterogeneity, impacting efficacy and safety, as well as increasing the risk of batch failure during manufacturing. To address this, we generated a novel analytical method to measure vector copy distribution at the single-cell level in a gene-modified, lentiviral-based immunotherapy model. As the limited amount of genomic DNA in a single cell hinders reliable quantification, we implemented a preamplification strategy on selected lentiviral and human gene targets in isolated live single cells, followed by quantification of amplified material by droplet digital PCR. Using a bespoke probability framework based on Bayesian statistics, we show that we can estimate vector copy number (VCN) integers with maximum likelihood scores. Notably, single-cell data are consistent with population analysis and also provide an overall measurement of transduction efficiency by discriminating transduced (VCN ≥ 1) from nontransduced (VCN = 0) cells. The ability to characterize cell-to-cell variability provides a powerful high-resolution approach for product characterization, which could ultimately allow improved control over product quality and safety.
Collapse
Affiliation(s)
- Ilaria Santeramo
- Cell and Gene Therapy Catapult, 12th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom
| | - Marta Bagnati
- Cell and Gene Therapy Catapult, 12th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom
| | - Emily Jane Harvey
- Cell and Gene Therapy Catapult, 12th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom
| | - Enas Hassan
- Cell and Gene Therapy Catapult, 12th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom
| | - Beata Surmacz-Cordle
- Cell and Gene Therapy Catapult, 12th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom
| | - Damian Marshall
- Cell and Gene Therapy Catapult, 12th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom
| | - Vincenzo Di Cerbo
- Cell and Gene Therapy Catapult, 12th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom
| |
Collapse
|
14
|
França TT, Barreiros LA, Al-Ramadi BK, Ochs HD, Cabral-Marques O, Condino-Neto A. CD40 ligand deficiency: treatment strategies and novel therapeutic perspectives. Expert Rev Clin Immunol 2019; 15:529-540. [PMID: 30681380 DOI: 10.1080/1744666x.2019.1573674] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION CD40 ligand (CD40L) deficiency or X-linked Hyper-IgM syndrome is a severe primary immunodeficiency caused by mutations in the CD40L gene. Despite currently available treatments, CD40L-deficient patients remain susceptible to life-threatening infections and have poor long term survival. Areas covered: Here, we discuss clinical and immunological characteristics of CD40L deficiency as well as current therapeutic strategies used for patient management. This review highlights that beyond B cell defects, patients' susceptibility to opportunistic pathogens might be due to impaired T cell and innate immune responses. In this context, we discuss how better knowledge of CD40L function and regulation may result in the development of new treatments. Expert opinion: Despite the introduction of hematopoietic stem-cell transplantation, immunoglobulin replacement, granulocyte colony-stimulating factor (G-CSF) administration, and prophylactic antibiotic therapies, life-threatening infections still cause high morbidity and mortality among CD40L-deficient patients. The reasons for this inadequate response to current therapies remains poorly understood, but recent reports suggest the involvement of CD40L-CD40 interaction in early stages of the innate immune system ontogeny. The development of novel gene therapeutic approaches and the use of redirected immunotherapies represent alternative treatment methods that could offer reduced morbidity and mortality rates for patients with CD40L deficiency.
Collapse
Affiliation(s)
- Tabata T França
- a Department of Immunology, Institute of Biomedical Sciences , University of São Paulo , São Paulo , Brazil
| | - Lucila A Barreiros
- a Department of Immunology, Institute of Biomedical Sciences , University of São Paulo , São Paulo , Brazil
| | - Basel K Al-Ramadi
- b Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences , UAE University , Al Ain , United Arab Emirates
| | - Hans D Ochs
- c Department of Pediatrics , University of Washington School of Medicine, and Seattle Children's Research Institute , Seattle , WA , USA
| | - Otavio Cabral-Marques
- d Department of Rheumatology and Clinical Immunology, Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, Faculty of Medicine , University of Freiburg , Freiburg , Germany
| | - Antonio Condino-Neto
- a Department of Immunology, Institute of Biomedical Sciences , University of São Paulo , São Paulo , Brazil
| |
Collapse
|
15
|
Mendoza-Torres E, Riquelme JA, Vielma A, Sagredo AR, Gabrielli L, Bravo-Sagua R, Jalil JE, Rothermel BA, Sanchez G, Ocaranza MP, Lavandero S. Protection of the myocardium against ischemia/reperfusion injury by angiotensin-(1–9) through an AT2R and Akt-dependent mechanism. Pharmacol Res 2018; 135:112-121. [DOI: 10.1016/j.phrs.2018.07.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/29/2018] [Accepted: 07/22/2018] [Indexed: 01/01/2023]
|
16
|
Rein LA, Yang H, Chao NJ. Applications of Gene Editing Technologies to Cellular Therapies. Biol Blood Marrow Transplant 2018; 24:1537-1545. [DOI: 10.1016/j.bbmt.2018.03.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 03/23/2018] [Indexed: 12/26/2022]
|
17
|
Lyu L, Hu M, Fu A, Xing B. Extracellular Vesicle Directed Exogenous Ion Channel Transport for Precise Manipulation of Biological Events. Bioconjug Chem 2018; 29:2715-2722. [DOI: 10.1021/acs.bioconjchem.8b00377] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Linna Lyu
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University, Singapore, 637371
| | - Ming Hu
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University, Singapore, 637371
| | - Afu Fu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore, 637459
| | - Bengang Xing
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University, Singapore, 637371
| |
Collapse
|
18
|
Miesbach W, Sawyer EK. Practical Implications of Factor IX Gene Transfer for Individuals with Hemophilia B: A Clinical Perspective. HUM GENE THER CL DEV 2018; 29:80-89. [PMID: 29624465 DOI: 10.1089/humc.2017.253] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Gene therapy for severe hemophilia is on the cusp of entering clinical practice. However, there is limited clinical experience in this area given that gene transfer is a relatively recent technology. Therefore, this clinical perspective article will review the evidence supporting gene therapy in this field, examine ways to open a dialogue about gene therapy with patients in the clinic setting, and present a case of a participant in a recent clinical trial of gene therapy for hemophilia. Clinical trials in hemophilia using adeno-associated virus (AAV) vectors to transfer functional factor IX (FIX) have reported increases in FIX activity to functional levels, reduced bleed frequency, and a lessening or abrogation of the need for costly FIX replacement. The safety profile of AAV-mediated gene therapy also appears positive, with manageable, asymptomatic increases in liver enzymes being the most commonly described adverse event. Examining a clinical case in hemophilia B more closely, gene transfer decreased annualized bleeds from six (unknown or spontaneous) bleeds before treatment to three (spontaneous) bleeds after treatment alongside a 55% reduction in FIX replacement. The participant experienced an increase in traumatic bleeds after treatment, which appears to reflect increased physical activity and early prophylaxis discontinuation. After the gene transfer, the participant considered his hemophilia to be "cured," which emphasizes the need to manage patient expectations, particularly regarding activity levels and bleed risk in the immediate post-treatment period. Gene therapy for hemophilia has the potential to transform the lives of affected individuals and is likely to create a new class of hemophilia patient who has shifted from a severe to a mild phenotype. Despite having a mild phenotype, these individuals may retain a legacy of increased bleed risk and joint damage from their years with severe hemophilia and will need different clinical management compared to a more typical individual with mild hemophilia.
Collapse
|
19
|
Yang Y, Hong Y, Cho E, Kim GB, Kim IS. Extracellular vesicles as a platform for membrane-associated therapeutic protein delivery. J Extracell Vesicles 2018; 7:1440131. [PMID: 29535849 PMCID: PMC5844050 DOI: 10.1080/20013078.2018.1440131] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 02/07/2018] [Indexed: 02/08/2023] Open
Abstract
Membrane proteins are of great research interest, particularly because they are rich in targets for therapeutic application. The suitability of various membrane proteins as targets for therapeutic formulations, such as drugs or antibodies, has been studied in preclinical and clinical studies. For therapeutic application, however, a protein must be expressed and purified in as close to its native conformation as possible. This has proven difficult for membrane proteins, as their native conformation requires the association with an appropriate cellular membrane. One solution to this problem is to use extracellular vesicles as a display platform. Exosomes and microvesicles are membranous extracellular vesicles that are released from most cells. Their membranes may provide a favourable microenvironment for membrane proteins to take on their proper conformation, activity, and membrane distribution; moreover, membrane proteins can cluster into microdomains on the surface of extracellular vesicles following their biogenesis. In this review, we survey the state-of-the-art of extracellular vesicle (exosome and small-sized microvesicle)-based therapeutics, evaluate the current biological understanding of these formulations, and forecast the technical advances that will be needed to continue driving the development of membrane protein therapeutics.
Collapse
Affiliation(s)
- Yoosoo Yang
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division for Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, Republic of Korea
| | - Yeonsun Hong
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Eunji Cho
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Gi Beom Kim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - In-San Kim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| |
Collapse
|
20
|
Mehta RS, Rezvani K. Chimeric Antigen Receptor Expressing Natural Killer Cells for the Immunotherapy of Cancer. Front Immunol 2018; 9:283. [PMID: 29497427 PMCID: PMC5818392 DOI: 10.3389/fimmu.2018.00283] [Citation(s) in RCA: 204] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 01/31/2018] [Indexed: 12/12/2022] Open
Abstract
Adoptive cell therapy has emerged as a powerful treatment for advanced cancers resistant to conventional agents. Most notable are the remarkable responses seen in patients receiving autologous CD19-redirected chimeric antigen receptor (CAR) T cells for the treatment of B lymphoid malignancies; however, the generation of autologous products for each patient is logistically cumbersome and has restricted widespread clinical use. A banked allogeneic product has the potential to overcome these limitations, yet allogeneic T-cells (even if human leukocyte antigen-matched) carry a major risk of graft-versus-host disease (GVHD). Natural killer (NK) cells are bone marrow-derived innate lymphocytes that can eliminate tumors directly, with their activity governed by the integration of signals from activating and inhibitory receptors and from cytokines including IL-15, IL-12, and IL-18. NK cells do not cause GVHD or other alloimmune or autoimmune toxicities and thus, can provide a potential source of allogeneic “off-the-shelf” cellular therapy, mediating major anti-tumor effects without inducing potentially lethal alloreactivity such as GVHD. Given the multiple unique advantages of NK cells, researchers are now exploring the use of CAR-engineered NK cells for the treatment of various hematological and non-hematological malignancies. Herein, we review preclinical data on the development of CAR-NK cells, advantages, disadvantages, and current obstacles to their clinical use.
Collapse
|
21
|
Alvarez-Urena P, Zhu B, Henslee G, Sonnet C, Davis E, Sevick-Muraca E, Davis A, Olmsted-Davis E. Development of a Cell-Based Gene Therapy Approach to Selectively Turn Off Bone Formation. J Cell Biochem 2017. [PMID: 28621436 DOI: 10.1002/jcb.26220] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cell and gene therapy approaches are safer when they possess a system that enables the therapy to be rapidly halted. Human mesenchymal stem cells were transduced with an adenoviral vector containing the cDNA for bone morphogenetic protein 2 (AdBMP2) to induce bone formation. To make this method safer, a system to quickly kill these virally transduced cells was designed and evaluated. Cells were encapsulated inside poly(ethylene glycol) diacrylate (PEG-Da) hydrogels that are able to shield the cells from immunological destruction. The system involves an inducible caspase-9 (iCasp9) activated using a specific chemical inducer of dimerization (CID). Delivering AdBMP2-transduced human mesenchymal stem cells encapsulated in PEG-Da hydrogel promoted ectopic ossification in vivo, and the iCasp9 system allowed direct control of the timing of apoptosis of the injected cells. The iCasp9-CID system enhances the safety of delivering AdBMP2-transduced cells, making it a more compelling therapeutic for bone repair and spine fusion. J. Cell. Biochem. 118: 3627-3634, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Pedro Alvarez-Urena
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas
| | - Banghe Zhu
- Center for Molecular Imaging, University of Texas Health Sciences Center, Houston, Texas.,Department of Pediatrics-Section Hematology/Oncology, Baylor College of Medicine, Houston, Texas
| | - Gabrielle Henslee
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas
| | - Corinne Sonnet
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas
| | - Eleanor Davis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas
| | - Eva Sevick-Muraca
- Center for Molecular Imaging, University of Texas Health Sciences Center, Houston, Texas
| | - Alan Davis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas.,Department of Pediatrics-Section Hematology/Oncology, Baylor College of Medicine, Houston, Texas.,Department of Orthopedic Surgery, Baylor College of Medicine, Houston, Texas
| | - Elizabeth Olmsted-Davis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas.,Department of Pediatrics-Section Hematology/Oncology, Baylor College of Medicine, Houston, Texas.,Department of Orthopedic Surgery, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
22
|
Igarashi Y, Uchiyama T, Minegishi T, Takahashi S, Watanabe N, Kawai T, Yamada M, Ariga T, Onodera M. Single Cell-Based Vector Tracing in Patients with ADA-SCID Treated with Stem Cell Gene Therapy. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017. [PMID: 28626778 PMCID: PMC5466583 DOI: 10.1016/j.omtm.2017.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Clinical improvement in stem cell gene therapy (SCGT) for primary immunodeficiencies depends on the engraftment levels of genetically corrected cells, and tracing the transgene in each hematopoietic lineage is therefore extremely important in evaluating the efficacy of SCGT. We established a single cell-based droplet digital PCR (sc-ddPCR) method consisting of the encapsulation of a single cell into each droplet, followed by emulsion PCR with primers and probes specific for the transgene. A fluorescent signal in a droplet indicates the presence of a single cell carrying the target gene in its genome, and this system can clearly determine the ratio of transgene-positive cells in the entire population at the genomic level. Using sc-ddPCR, we analyzed the engraftment of vector-transduced cells in two patients with severe combined immunodeficiency (SCID) who were treated with SCGT. Sufficient engraftment of the transduced cells was limited to the T cell lineage in peripheral blood (PB), and a small percentage of CD34+ cells exhibited vector integration in bone marrow, indicating that the transgene-positive cells in PB might have differentiated from a small population of stem cells or lineage-restricted precursor cells. sc-ddPCR is a simplified and powerful tool for the detailed assessment of transgene-positive cell distribution in patients treated with SCGT.
Collapse
Affiliation(s)
- Yuka Igarashi
- Department of Human Genetics, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Toru Uchiyama
- Department of Human Genetics, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Tomoko Minegishi
- Department of Human Genetics, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Sirirat Takahashi
- Department of Human Genetics, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Nobuyuki Watanabe
- Department of Human Genetics, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Toshinao Kawai
- Department of Human Genetics, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Masafumi Yamada
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, Hokkaido 060-8638, Japan
| | - Tadashi Ariga
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, Hokkaido 060-8638, Japan
| | - Masafumi Onodera
- Department of Human Genetics, National Center for Child Health and Development, Tokyo 157-8535, Japan
| |
Collapse
|
23
|
Yucel D, Kocabas F. Developments in Hematopoietic Stem Cell Expansion and Gene Editing Technologies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1079:103-125. [DOI: 10.1007/5584_2017_114] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
24
|
The Functionality of Minimal PiggyBac Transposons in Mammalian Cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2016; 5:e369. [PMID: 27701401 PMCID: PMC5095681 DOI: 10.1038/mtna.2016.76] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 07/12/2016] [Indexed: 12/11/2022]
Abstract
Minimal piggyBac vectors are a modified single-plasmid version of the classical piggyBac delivery system that can be used for stable transgene integration. These vectors have a truncated terminal domain in the delivery cassette and thus, integrate significantly less flanking transposon DNA into host cell chromatin than classical piggyBac vectors. Herein, we test various characteristics of this modified transposon. The integration efficiency of minimal piggyBac vectors was inversely related to the size of both the transposon and the entire plasmid, but inserts as large as 15 kb were efficiently integrated. Open and super-coiled vectors demonstrated the same integration efficiency while DNA methylation decreased the integration efficiency and silenced the expression of previously integrated sequences in some cell types. Importantly, the incidence of plasmid backbone integration was not increased above that seen in nontransposon control vectors. In BALB/c mice, we demonstrated prolonged expression of two transgenes (intracellular mCherry and secretable Gaussia luciferase) when delivered by the minimal piggyBac that resulted in a more sustained antibody production against the immunogenic luciferase than when delivered by a transient (nontransposon) vector plasmid. We conclude that minimal piggyBac vectors are an effective alternative to other integrative systems for stable DNA delivery in vitro and in vivo.
Collapse
|
25
|
Casey NP, Fujiwara H, Tanimoto K, Okamoto S, Mineno J, Kuzushima K, Shiku H, Yasukawa M. A Functionally Superior Second-Generation Vector Expressing an Aurora Kinase-A-Specific T-Cell Receptor for Anti-Leukaemia Adoptive Immunotherapy. PLoS One 2016; 11:e0156896. [PMID: 27271876 PMCID: PMC4896450 DOI: 10.1371/journal.pone.0156896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 05/21/2016] [Indexed: 11/23/2022] Open
Abstract
Aurora Kinase A is a cancer-associated protein normally involved in the regulation of mitosis. Being over-expressed in a range of cancers, it is a suitable target for cell-based immunotherapy. Gene transfer of T-cell receptor sequences cognisant of HLA-A*0201-restricted Aurora Kinase A antigen has previously been shown to transfer specific immunoreactivity against the target peptide in a Human Lymphocyte Antigen-restricted manner. While T cell receptor gene-transfer has great potential in overcoming the difficulties of isolating and expanding tumour-reactive lymphocytes from a patient’s own cells, one hurdle is potential mispairing and competition between exogenous and endogenous T cell receptor chains. We have used a retroviral vector design bearing a short-interfering RNA that downregulates endogenous T cell receptor chains, without affecting expression of the transgenic T cell receptor sequences. The T cell receptor expression cassette also includes a 2A self-cleaving peptide, resulting in equimolar expression of the T cell receptor alpha and beta chains, further enhancing formation of the desired T cell receptor. Via a simple, modular cloning method, we have cloned the alpha and beta chains of the anti-Aurora Kinase A-reactive T cell receptor into this ‘siTCR’ vector. We then compared the activity of this vector against the original, ‘conventional’ vector across a panel of assays. T cell receptors expressed from the siTCR-vector retained the cytotoxic functionality of the original vector, with evidence of reduced off-target reactivity. The rate of expression of correctly-formed T cell receptors was superior using the siTCR design, and this was achieved at lower vector copy numbers. Maintaining T cell receptor efficacy with a reduced vector copy number reduces the risk of genotoxicity. The siTCR design also reduces the risk of mispairing and cross-reactivity, while increasing the functional titre. Such improvements in the safety of T cell receptor gene-transfer will be crucial for clinical applications of this technology.
Collapse
Affiliation(s)
- Nicholas Paul Casey
- Department of Hematology, Clinical Immunology and Infectious Disease, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Hiroshi Fujiwara
- Department of Hematology, Clinical Immunology and Infectious Disease, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Kazushi Tanimoto
- Department of Hematology, Clinical Immunology and Infectious Disease, Ehime University Graduate School of Medicine, Ehime, Japan
| | | | | | | | - Hiroshi Shiku
- Department of Cancer Vaccine and Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie, Japan
| | - Masaki Yasukawa
- Department of Hematology, Clinical Immunology and Infectious Disease, Ehime University Graduate School of Medicine, Ehime, Japan
| |
Collapse
|
26
|
Cortese FAB, Santostasi G. Whole-Body Induced Cell Turnover: A Proposed Intervention for Age-Related Damage and Associated Pathology. Rejuvenation Res 2016; 19:322-36. [PMID: 26649945 DOI: 10.1089/rej.2015.1763] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
In both biomedicine in general and biomedical gerontology in particular, cell replacement therapy is traditionally proposed as an intervention for cell loss. This article presents a proposed intervention-whole-body induced cell turnover (WICT)-for use in biomedical gerontology that combines cell replacement therapy with a second therapeutic component (targeted cell ablation) so as to broaden the therapeutic utility of cell therapies and increase the categories of age-related damage that are amenable to cell-based interventions. In particular, WICT may allow cell therapies to serve as an intervention for accumulated cellular and intracellular damage, such as telomere depletion, genomic DNA and mitochondrial DNA damage and mutations, replicative senescence, functionally deleterious age-related changes in gene expression, accumulated cellular and intracellular aggregates, and functionally deleterious posttranslationally modified gene products. WICT consists of the gradual ablation and subsequent replacement of a patient's entire set of constituent cells gradually over the course of their adult life span through the quantitative and qualitative coordination of targeted cell ablation with exogenous cell administration. The aim is to remove age-associated cellular and intracellular damage present in the patient's endogenous cells. In this study, we outline the underlying techniques and technologies by which WICT can be mediated, describe the mechanisms by which it can serve to negate or prevent age-related cellular and intracellular damage, explicate the unique therapeutic components and utilities that distinguish it as a distinct type of cell-based intervention for use in biomedical gerontology, and address potential complications associated with the therapy.
Collapse
Affiliation(s)
| | - Giovanni Santostasi
- 2 Department of Neurology, Feinberg School of Medicine, Northwestern University , Chicago, Illinois
| |
Collapse
|
27
|
Daniel MG, Pereira CF, Lemischka IR, Moore KA. Making a Hematopoietic Stem Cell. Trends Cell Biol 2015; 26:202-214. [PMID: 26526106 DOI: 10.1016/j.tcb.2015.10.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 09/29/2015] [Accepted: 10/01/2015] [Indexed: 12/22/2022]
Abstract
Previous attempts to either generate or expand hematopoietic stem cells (HSCs) in vitro have involved either ex vivo expansion of pre-existing patient or donor HSCs or de novo generation from pluripotent stem cells (PSCs), comprising both embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). iPSCs alleviated ESC ethical issues but attempts to generate functional mature hematopoietic stem and progenitor cells (HSPCs) have been largely unsuccessful. New efforts focus on directly reprogramming somatic cells into definitive HSCs and HSPCs. To meet clinical needs and to advance drug discovery and stem cell therapy, alternative approaches are necessary. In this review, we synthesize the strategies used and the key findings made in recent years by those trying to make an HSC.
Collapse
Affiliation(s)
- Michael G Daniel
- Department of Developmental and Regenerative Biology, Icahn School of Medicine, New York, NY, USA; Black Family Stem Cell Institute, Icahn School of Medicine, New York, NY, USA; The Graduate School of Biomedical Science, Icahn School of Medicine, New York, NY, USA
| | - Carlos-Filipe Pereira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC Biotech Building, Biocant Park, 3060-197 Cantanhede, Portugal
| | - Ihor R Lemischka
- Department of Developmental and Regenerative Biology, Icahn School of Medicine, New York, NY, USA; Black Family Stem Cell Institute, Icahn School of Medicine, New York, NY, USA; Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine, New York, NY, USA
| | - Kateri A Moore
- Department of Developmental and Regenerative Biology, Icahn School of Medicine, New York, NY, USA; Black Family Stem Cell Institute, Icahn School of Medicine, New York, NY, USA.
| |
Collapse
|
28
|
Vu LT, Nguyen TTK, Alam S, Sakamoto T, Fujimoto K, Suzuki H, Tsukahara T. Changing blue fluorescent protein to green fluorescent protein using chemical RNA editing as a novel strategy in genetic restoration. Chem Biol Drug Des 2015; 86:1242-52. [PMID: 26031895 DOI: 10.1111/cbdd.12592] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 04/15/2015] [Accepted: 05/14/2015] [Indexed: 11/29/2022]
Abstract
Using the transition from cytosine of BFP (blue fluorescent protein) gene to uridine of GFP (green fluorescent protein) gene at position 199 as a model, we successfully controlled photochemical RNA editing to effect site-directed deamination of cytidine (C) to uridine (U). Oligodeoxynucleotides (ODNs) containing 5'-carboxyvinyl-2'-deoxyuridine ((CV) U) were used for reversible photoligation, and single-stranded 100-nt BFP DNA and in vitro-transcribed full-length BFP mRNA were the targets. Photo-cross-linking with the responsive ODNs was performed using UV (366 nm) irradiation, which was followed by heat treatment, and the cross-linked nucleotide was cleaved through photosplitting (UV, 312 nm). The products were analyzed using restriction fragment length polymorphism (RFLP) and fluorescence measurements. Western blotting and fluorescence-analysis results revealed that in vitro-translated proteins were synthesized from mRNAs after site-directed RNA editing. We detected substantial amounts of the target-base-substituted fragment using RFLP and observed highly reproducible spectra of the transition-GFP signal using fluorescence spectroscopy, which indicated protein stability. ODNc restored approximately 10% of the C-to-U transition. Thus, we successfully used non-enzymatic site-directed deamination for genetic restoration in vitro. In the near future, in vivo studies that include cultured cells and model animals will be conducted to treat genetic disorders.
Collapse
Affiliation(s)
- Luyen T Vu
- School of Materials Science, Japan Advanced Institute of Science and Technology (JAIST), 1-1 Asahidai, Nomi City, Ishikawa, 923-1292, Japan
| | - Thanh T K Nguyen
- School of Materials Science, Japan Advanced Institute of Science and Technology (JAIST), 1-1 Asahidai, Nomi City, Ishikawa, 923-1292, Japan
| | - Shafiul Alam
- School of Materials Science, Japan Advanced Institute of Science and Technology (JAIST), 1-1 Asahidai, Nomi City, Ishikawa, 923-1292, Japan
| | - Takashi Sakamoto
- School of Materials Science, Japan Advanced Institute of Science and Technology (JAIST), 1-1 Asahidai, Nomi City, Ishikawa, 923-1292, Japan
| | - Kenzo Fujimoto
- School of Materials Science, Japan Advanced Institute of Science and Technology (JAIST), 1-1 Asahidai, Nomi City, Ishikawa, 923-1292, Japan
| | - Hitoshi Suzuki
- School of Materials Science, Japan Advanced Institute of Science and Technology (JAIST), 1-1 Asahidai, Nomi City, Ishikawa, 923-1292, Japan
| | - Toshifumi Tsukahara
- School of Materials Science, Japan Advanced Institute of Science and Technology (JAIST), 1-1 Asahidai, Nomi City, Ishikawa, 923-1292, Japan
| |
Collapse
|
29
|
ZHOU HAN, MA XIAOFENG, LIU YONGZE, DONG LEI, LUO YI, ZHU GUANGJIE, QIAN XIAOYUN, CHEN JIE, LU LIN, WANG JUNGUO, GAO XIA. Linear polyethylenimine-plasmid DNA nanoparticles are ototoxic to the cultured sensory epithelium of neonatal mice. Mol Med Rep 2015; 11:4381-8. [DOI: 10.3892/mmr.2015.3306] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 01/15/2015] [Indexed: 11/06/2022] Open
|
30
|
Antibody-modified T cells: CARs take the front seat for hematologic malignancies. Blood 2014; 123:2625-35. [PMID: 24578504 DOI: 10.1182/blood-2013-11-492231] [Citation(s) in RCA: 473] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
T cells redirected to specific antigen targets with engineered chimeric antigen receptors (CARs) are emerging as powerful therapies in hematologic malignancies. Various CAR designs, manufacturing processes, and study populations, among other variables, have been tested and reported in over 10 clinical trials. Here, we review and compare the results of the reported clinical trials and discuss the progress and key emerging factors that may play a role in effecting tumor responses. We also discuss the outlook for CAR T-cell therapies, including managing toxicities and expanding the availability of personalized cell therapy as a promising approach to all hematologic malignancies. Many questions remain in the field of CAR T cells directed to hematologic malignancies, but the encouraging response rates pave a wide road for future investigation.
Collapse
|
31
|
Abstract
INTRODUCTION Cardiovascular gene therapy is the third most popular application for gene therapy, representing 8.4% of all gene therapy trials as reported in 2012 estimates. Gene therapy in cardiovascular disease is aiming to treat heart failure from ischemic and non-ischemic causes, peripheral artery disease, venous ulcer, pulmonary hypertension, atherosclerosis and monogenic diseases, such as Fabry disease. AREAS COVERED In this review, we will focus on elucidating current molecular targets for the treatment of ventricular dysfunction following myocardial infarction (MI). In particular, we will focus on the treatment of i) the clinical consequences of it, such as heart failure and residual myocardial ischemia and ii) etiological causes of MI (coronary vessels atherosclerosis, bypass venous graft disease, in-stent restenosis). EXPERT OPINION We summarise the scheme of the review and the molecular targets either already at the gene therapy clinical trial phase or in the pipeline. These targets will be discussed below. Following this, we will focus on what we believe are the 4 prerequisites of success of any gene target therapy: safety, expression, specificity and efficacy (SESE).
Collapse
Affiliation(s)
- Maria C Scimia
- Temple University, Translational Medicine/Pharmacology , 3500 N. Broad Street, Philadelphia, 19140 , USA
| | | | | |
Collapse
|
32
|
Scherzed A, Hackenberg S, Radeloff A, Froelich K, Rak K, Hagen R, Kleinsasser N. Human mesenchymal stem cells promote cancer motility and cytokine secretion in vitro. Cells Tissues Organs 2013; 198:327-37. [PMID: 24356396 DOI: 10.1159/000356731] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2013] [Indexed: 11/19/2022] Open
Abstract
Interactions of human mesenchymal stem cells (hMSC) with tumors are controversially discussed since there is evidence for both tumor progression as well as tumor inhibition by hMSC. The objective of the present study is to investigate whether hMSC support cell motility and cytokine secretion in a head and neck squamous cell carcinoma cell line (HLaC 78). A spheroid model was generated in which the ultrastructure of spheroids was analyzed using scanning electron microscopy (SEM). The migration capability was monitored in a monolayer as well as in a spheroid model. The variation in migration and secretion of interleukin (IL)-6, IL-8 and vascular endothelial growth factor (VEGF), as well as the expression of the multidrug resistance gene (MDR-1) was investigated. Finally, the alteration in the cell cycle was analyzed by flow cytometry. SEM showed a tight cell-cell contact with extensive secretion of extracellular matrix. The migration and invasion capability of HLaC 78 was enhanced by hMSC. Cancer cell motility was also increased by hMSC as well as secretion of the cytokines IL-6, IL-8 and VEGF. hMSC did not induce the expression of MDR-1 in HLaC 78, and there was no alteration in the cell cycle of HLaC 78 after cocultivation with hMSC. Our results confirm the important role of hMSC in cancer biology since both an enhancement of cell motility as well as cytokine secretion could be shown. However, based on these findings and those in the current literature, caution must be applied when using hMSC as a carrier for tumor therapy in cancer treatment.
Collapse
Affiliation(s)
- A Scherzed
- Department of Otorhinolaryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, University Hospital of Würzburg, Würzburg, Germany
| | | | | | | | | | | | | |
Collapse
|
33
|
Iwami K, Natsume A, Ohno M, Ikeda H, Mineno J, Nukaya I, Okamoto S, Fujiwara H, Yasukawa M, Shiku H, Wakabayashi T. Adoptive transfer of genetically modified Wilms' tumor 1-specific T cells in a novel malignant skull base meningioma model. Neuro Oncol 2013; 15:747-58. [PMID: 23460320 DOI: 10.1093/neuonc/not007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Meningiomas are the most commonly diagnosed primary intracranial neoplasms. Despite significant advances in modern therapies, the management of malignant meningioma and skull base meningioma remains a challenge. Thus, the development of new treatment modalities is urgently needed for these difficult-to-treat meningiomas. The goal of this study was to investigate the potential of build-in short interfering RNA-based Wilms' tumor protein (WT1)-targeted adoptive immunotherapy in a reproducible mouse model of malignant skull base meningioma that we recently established. METHODS We compared WT1 mRNA expression in human meningioma tissues and gliomas by quantitative real-time reverse-transcription polymerase chain reaction. Human malignant meningioma cells (IOMM-Lee cells) were labeled with green fluorescent protein (GFP) and implanted at the skull base of immunodeficient mice by using the postglenoid foramen injection (PGFi) technique. The animals were sacrificed at specific time points for analysis of tumor formation. Two groups of animals received adoptive immunotherapy with control peripheral blood mononuclear cells (PBMCs) or WT1-targeted PBMCs. RESULTS High levels of WT1 mRNA expression were observed in many meningioma tissues and all meningioma cell lines. IOMM-Lee-GFP cells were successfully implanted using the PGFi technique, and malignant skull base meningiomas were induced in all mice. The systemically delivered WT1-targeted PBMCs infiltrated skull base meningiomas and significantly delayed tumor growth and increased survival time. CONCLUSIONS We have established a reproducible mouse model of malignant skull base meningioma. WT1-targeted adoptive immunotherapy appears to be a promising approach for the treatment of difficult-to-treat meningiomas.
Collapse
Affiliation(s)
- Kenichiro Iwami
- Department of Neurosurgery, Nagoya University, Graduate School of Medicine, 65, Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
A Promising Vector for TCR Gene Therapy: Differential Effect of siRNA, 2A Peptide, and Disulfide Bond on the Introduced TCR Expression. MOLECULAR THERAPY. NUCLEIC ACIDS 2012; 1:e63. [PMID: 23250361 PMCID: PMC3528300 DOI: 10.1038/mtna.2012.52] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Adoptive immunotherapy using TCR gene-modified T-lymphocytes is an attractive strategy for targeting malignancies. However, TCR mispairings between endogenous and introduced TCR chains are a major concern, as they may induce mixed TCRs with unknown specificities and may reduce the expression of therapeutic TCRs. To overcome these problems, we have recently established a novel retroviral siTCR vector encoding small-interfering RNAs (siRNAs) to knockdown endogenous TCR genes for the efficient expression of therapeutic TCRs. In this study, to improve the efficacy of siTCR vectors, we developed 2A peptide-based siTCR vectors that could increase the expression levels of transduced TCRs compared with internal promoter-based siTCR vectors. We also evaluated the efficacy of an siTCR strategy and the addition of a new interchain disulfide bond created by cysteine modification. We found that the effect of the cysteine modification depended on TCR variations, while the siTCR strategy improved the expression of all TCRs tested. Furthermore, the combined effect of the siTCR and cysteine modification strategies was highly significant for certain TCRs. Therefore, our novel siTCR technology, in isolation or in combination with another strategy, may open the door to effective immunotherapy for cancer patients.Molecular Therapy - Nucleic Acids (2012) 1, e63. doi:10.1038/mtna.2012.52; published online 18 December 2012.
Collapse
|
35
|
Sewbalas A, Islam RU, van Otterlo WAL, de Koning CB, Singh M, Arbuthnot P, Ariatti M. Enhancement of transfection activity in HEK293 cells by lipoplexes containing cholesteryl nitrogen-pivoted aza-crown ethers. Med Chem Res 2012. [DOI: 10.1007/s00044-012-0252-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
36
|
Abstract
Stable cell lines of Chinese hamster ovary (CHO) cells are the predominant source of commercial -biopharmaceutical proteins. Because making suitable CHO cell lines is time-consuming and costly, -preliminary experiments with transient expression are usually performed to optimize as many protein -production parameters as possible. Here, we describe protocols for optimizing expression in transient expression experiments and isolating stable CHO cell lines using two types of self-made reagents, namely, lipoplexes and polyplexes.
Collapse
Affiliation(s)
- Renate Kunert
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria.
| | | |
Collapse
|
37
|
Abstract
Substantial advances in our understanding of lentivirus lifecycles and their various constituent proteins have permitted the bioengineering of lentiviral vectors now considered safe enough for clinical trials for both lethal and non-lethal diseases. They possess distinct properties that make them particularly suitable for gene delivery in ophthalmic diseases, including high expression, consistent targeting of various post-mitotic ocular cells in vivo and a paucity of associated intraocular inflammation, all contributing to their ability to mediate efficient and stable intraocular gene transfer. In this review, the intraocular tropisms and therapeutic applications of both primate and non-primate lentiviral vectors, and how the unique features of the eye influence these, are discussed. The feasibility of therapeutic targeting using these vectors in animal models of both anterior and posterior ophthalmic disorders has been established, and has, in combination with substantial progress in enhancing lentiviral vector bio-safety over the past two decades, paved the way for the first human ophthalmic clinical trials using lentivirus-based gene transfer vectors.
Collapse
Affiliation(s)
- K S Balaggan
- Department of Genetics, Institute of Ophthalmology, London, UK.
| | | |
Collapse
|
38
|
Mattar CN, Choolani M, Biswas A, Waddington SN, Chan JKY. Fetal gene therapy: recent advances and current challenges. Expert Opin Biol Ther 2011; 11:1257-71. [PMID: 21623703 DOI: 10.1517/14712598.2011.585153] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Fetal gene therapy (FGT) can potentially be applied to perinatally lethal monogenic diseases for rescuing clinically severe phenotypes, increasing the probability of intact neurological and other key functions at birth, or inducing immune tolerance to a transgenic protein to facilitate readministration of the vector/protein postnatally. As the field is still at an experimental stage, there are several important considerations regarding the practicality and the ethics of FGT. AREAS COVERED Here, through a review of FGT studies, the authors discuss the role and applications of FGT, the progress made with animal models that simulate human development, possible adverse effects in the recipient fetus and the mother and factors that affect clinical translation. EXPERT OPINION Although there are valid safety and ethical concerns, the authors argue that there may soon be enough convincing evidence from non-human primate models to take the next step towards clinical trials in the near future.
Collapse
Affiliation(s)
- Citra N Mattar
- Yong Loo Lin School of Medicine, National University of Singapore, Department of Obstetrics and Gynaecology, Experimental Fetal Medicine Group, NUHS Tower Block, Level 12, 1E Kent Ridge Road, 119228 Singapore
| | | | | | | | | |
Collapse
|
39
|
Abstract
The β-thalassaemias are inherited anaemias that form the most common class of monogenic disorders in the world. Treatment options are limited, with allogeneic haematopoietic stem cell transplantation offering the only hope for lifelong cure. However, this option is not available for many patients as a result of either the lack of compatible donors or the increased risk of transplant-related mortality in subjects with organ damage resulting from accumulated iron. The paucity of alternative treatments for patients that fall into either of these categories has led to the development of a revolutionary treatment strategy based on gene therapy. This approach involves replacing allogeneic stem cell transplantation with the transfer of normal globin genes into patient-derived, autologous haematopoietic stem cells. This highly attractive strategy offers several advantages, including bypassing the need for allogeneic donors and the immunosuppression required to achieve engraftment of the transplanted cells and to eliminate the risk of donor-related graft-versus-host disease. This review discusses the many advances that have been made towards this endeavour as well as the hurdles that must still be overcome before gene therapy for β-thalassaemia, as well as many other gene therapy applications, can be widely applied in the clinic.
Collapse
|
40
|
Momin EN, Vela G, Zaidi HA, Quiñones-Hinojosa A. The Oncogenic Potential of Mesenchymal Stem Cells in the Treatment of Cancer: Directions for Future Research. ACTA ACUST UNITED AC 2010; 6:137-148. [PMID: 20490366 DOI: 10.2174/157339510791111718] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) represent a promising new approach to the treatment of several diseases that are associated with dismal outcomes. These include myocardial damage, graft versus host disease, and possibly cancer. Although the potential therapeutic aspects of MSCs continue to be well-researched, the possible hazards of MSCs, and in particular their oncogenic capacity are poorly understood. This review addresses the oncogenic and tumor-supporting potential of MSCs within the context of cancer treatment. The risk for malignant transformation is discussed for each stage of the clinical lifecycle of MSCs. This includes malignant transformation in vitro during production phases, during insertion of potentially therapeutic transgenes, and finally in vivo via interactions with tumor stroma. The immunosuppressive qualities of MSCs, which may facilitate evasion of the immune system by a tumor, are also addressed. Limitations of the methods employed in clinical trials to date are reviewed, including the absence of long term follow-up and lack of adequate screening methods to detect formation of new tumors. Through discussions of the possible oncogenic and tumor-supporting mechanisms of MSCs, directions for future research are identified which may eventually facilitate the future clinical translation of MSCs for the treatment of cancer and other diseases.
Collapse
Affiliation(s)
- Eric N Momin
- Department of Neurosurgery and Oncology, The Johns Hopkins School of Medicine, Baltimore, MD
| | | | | | | |
Collapse
|
41
|
Sorrell DA, Robinson CJ, Smith JA, Kolb AF. Recombinase mediated cassette exchange into genomic targets using an adenovirus vector. Nucleic Acids Res 2010; 38:e123. [PMID: 20371519 PMCID: PMC2887974 DOI: 10.1093/nar/gkq192] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Recombinase mediated cassette exchange (RMCE) is a process in which site-specific recombinases exchange one gene cassette flanked by a pair of incompatible target sites for another cassette flanked by an identical pair of sites. Typically one cassette is present in the host genome, whereas the other gene cassette is introduced into the host cell by chemical or biological means. We show here that the frequency of cassette exchange is dependent on the relative and absolute quantities of the transgene cassette and the recombinase. We were able to successfully modify genomic targets not only by electroporation or chemically mediated gene transfer but also by using an adenovirus vector carrying both the transgene cassette to be inserted and the recombinase coding region. RMCE proceeds efficiently in cells in which the adenovirus vector is able to replicate. In contrast, insufficient quantities of the transgene cassette are produced in cells in which the virus cannot replicate. Additional transfection of the transgene cassette significantly enhances the RMCE frequency. This demonstrates that an RMCE system in the context of a viral vector allows the site directed insertion of a transgene into a defined genomic site.
Collapse
Affiliation(s)
- David A Sorrell
- Molecular Recognition Group, Hannah Research Institute, Ayr, KA6 5HL, UK
| | | | | | | |
Collapse
|
42
|
Thieme S, Ryser M, Gentsch M, Navratiel K, Brenner S, Stiehler M, Rölfing J, Gelinsky M, Rösen-Wolff A. Stromal cell-derived factor-1alpha-directed chemoattraction of transiently CXCR4-overexpressing bone marrow stromal cells into functionalized three-dimensional biomimetic scaffolds. Tissue Eng Part C Methods 2010; 15:687-96. [PMID: 19260802 DOI: 10.1089/ten.tec.2008.0556] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Three-dimensional (3D) bone substitute material should not only serve as scaffold in large bone defects but also attract mesenchymal stem cells, a subset of bone marrow stromal cells (BMSCs) that are able to form new bone tissue. An additional crucial step is to attract BMSCs from the surface into deeper structures of 3D porous bone substitute scaffolds. Here we show that transient overexpression of CXCR4 in human BMSCs induced by mRNA transfection enhances stromal cell-derived factor-1alpha (SDF-1alpha)-directed chemotactic capacity to invade internal compartments of porous 3D bone substitute scaffolds in vitro and in vivo. In vitro native BMCSs invaded up to 500 mum into SDF-1alpha-releasing 3D scaffolds, whereas CXCR4-overexpressing BMSCs invaded up to 800 mum within 5 days. In addition, 60% downregulation of endogenous SDF-1 transcription in BMSCs by endoribonuclease-prepared siRNA before CXCR4 mRNA transfection enhanced SDF-1alpha-directed migration of human BMSCs by 50%. Implantation of SDF-1alpha-releasing scaffolds seeded with transiently CXCR4-overexpressing BMSCs resulted in an increase of invasion into internal compartments of the scaffolds in a mouse model. In vivo native BMCS invaded up to 250 mum into SDF-1alpha-releasing 3D scaffolds, whereas CXCR4-overexpressing BMSC invaded up to 500 mum within 5 days. Thus, the SDF-1alpha/CXCR4 chemoattraction system can be used to efficiently recruit BMSCs into SDF-1alpha-releasing 3D scaffolds in vitro and in vivo.
Collapse
Affiliation(s)
- Sebastian Thieme
- Department of Pediatrics, University Clinic Carl Gustav Carus, Technische Universität Dresden , Dresden, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Okamoto S, Mineno J, Ikeda H, Fujiwara H, Yasukawa M, Shiku H, Kato I. Improved expression and reactivity of transduced tumor-specific TCRs in human lymphocytes by specific silencing of endogenous TCR. Cancer Res 2009; 69:9003-11. [PMID: 19903853 DOI: 10.1158/0008-5472.can-09-1450] [Citation(s) in RCA: 149] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Adoptive T-cell therapy using lymphocytes genetically engineered to express tumor antigen-specific TCRs is an attractive strategy for treating patients with malignancies. However, there are potential drawbacks to this strategy: mispairing of the introduced TCR alpha/beta chains with the endogenous TCR subunits and competition of CD3 molecules between the introduced and endogenous TCRs can impair cell surface expression of the transduced TCR, resulting in insufficient function and potential generation of autoreactive T cells. In addition, the risk of tumor development following the infusion of cells with aberrant vector insertion sites increases with the vector copy number in the transduced cells. In this study, we developed retroviral vectors encoding both small interfering RNA constructs that specifically down-regulate endogenous TCR and a codon-optimized, small interfering RNA-resistant TCR specific for the human tumor antigens MAGE-A4 or WT1. At low copy numbers of the integrated vector, the transduced human lymphocytes exhibited high surface expression of the introduced tumor-specific TCR and reduced expression of endogenous TCRs. In consequence, the vector-transduced lymphocytes showed enhanced cytotoxic activity against antigen-expressing tumor cells. Therefore, our novel TCR gene therapy may open a new gate for effective immunotherapy in cancer patients.
Collapse
Affiliation(s)
- Sachiko Okamoto
- Center for Cell and Gene Therapy, Takara Bio, Inc., Shiga, Japan
| | | | | | | | | | | | | |
Collapse
|
44
|
Chang AH, Stephan MT, Lisowski L, Sadelain M. Erythroid-specific human factor IX delivery from in vivo selected hematopoietic stem cells following nonmyeloablative conditioning in hemophilia B mice. Mol Ther 2008; 16:1745-52. [PMID: 18682698 DOI: 10.1038/mt.2008.161] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
We have developed a lentiviral vector system for human factor IX (hFIX) gene transfer in hematopoietic stem cells (HSCs) that provides erythroid cell-derived systemic protein delivery following nonmyeloablative conditioning and in vivo methylguanine methyltransferase (MGMT) drug selection. After bone marrow transplantation under moderate Busulfan conditioning, the initial hFIX expression in the chimeras was minimally detectable. However, the hFIX levels rose sharply following in vivo MGMT-drug selection and eventually reached a level that is considered curative in hemophilia B therapy (>500 ng/ml). The rise of hFIX levels was proportional to the increase in vector copy (VC) number in peripheral blood cells. High levels of hFIX expression were maintained in serially engrafted mice chimeras for 18 months. Importantly, high-level hFIX expression by erythroid cells did not result in anemia or adversely affect red blood cell counts. The prospect of combining reduced intensity conditioning, a presumably lowered risk of insertional mutagenesis due to low VC number requirement and erythroid-restricted transgene expression, as well as long-term protein expression at high level, strongly supports the potential applicability of adult stem cell-based gene therapy in nonlethal blood or metabolic disorders, as demonstrated here for hemophilia.
Collapse
Affiliation(s)
- Alex H Chang
- Center for Cell Engineering, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
| | | | | | | |
Collapse
|
45
|
Lisowski L, Sadelain M. Current status of globin gene therapy for the treatment of β-thalassaemia. Br J Haematol 2008; 141:335-45. [DOI: 10.1111/j.1365-2141.2008.07098.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
46
|
Abstract
Highly active antiretroviral therapy prolongs the life of HIV-infected individuals, but it requires lifelong treatment and results in cumulative toxicities and viral-escape mutants. Gene therapy offers the promise of preventing progressive HIV infection by sustained interference with viral replication in the absence of chronic chemotherapy. Gene-targeting strategies are being developed with RNA-based agents, such as ribozymes, antisense, RNA aptamers and small interfering RNA, and protein-based agents, such as the mutant HIV Rev protein M10, fusion inhibitors and zinc-finger nucleases. Recent advances in T-cell-based strategies include gene-modified HIV-resistant T cells, lentiviral gene delivery, CD8(+) T cells, T bodies and engineered T-cell receptors. HIV-resistant hematopoietic stem cells have the potential to protect all cell types susceptible to HIV infection. The emergence of viral resistance can be addressed by therapies that use combinations of genetic agents and that inhibit both viral and host targets. Many of these strategies are being tested in ongoing and planned clinical trials.
Collapse
Affiliation(s)
- John J Rossi
- Division of Molecular Biology, Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, California 91010, USA.
| | | | | |
Collapse
|
47
|
Deschamps M, Mercier-Lethondal P, Certoux JM, Henry C, Lioure B, Pagneux C, Cahn JY, Deconinck E, Robinet E, Tiberghien P, Ferrand C. Deletions within the HSV-tk transgene in long-lasting circulating gene-modified T cells infused with a hematopoietic graft. Blood 2007; 110:3842-52. [PMID: 17717134 DOI: 10.1182/blood-2007-04-087346] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractIn our previous phase 1/2 study aimed at controlling graft-versus-host disease, 12 patients received Herpes simplex virus thymidine kinase (HSV-tk+)/neomycin phosphotransferase (NeoR+)–expressing donor gene-modified T cells (GMCs) and underwent an HLA-identical sibling T-cell–depleted bone marrow transplantation (BMT). This study's objective was to follow up, to quantify, and to characterize persistently circulating GMCs more than 10 years after BMT. Circulating GMCs remain detectable in all 4 evaluable patients. However, NeoR- and HSV-tk–polymerase chain reaction (PCR) differently quantified in vivo counts, suggesting deletions within the HSV-tk gene. Further experiments, including a novel “transgene walking” PCR method, confirmed the presence of deletions. The deletions were unique, patient-specific, present in most circulating GMCs expressing NeoR, and shown to occur at time of GMC production. Unique patient-specific retroviral insertion sites (ISs) were found in all GMCs capable of in vitro expansion/cloning as well. These findings suggest a rare initial gene deletion event and an in vivo survival advantage of rare GMC clones resulting from an anti–HSV-tk immune response and/or ganciclovir treatment. In conclusion, we show that donor mature T cells infused with a T-cell–depleted graft persist in vivo for more than a decade. These cells, containing transgene deletions and subjected to significant in vivo selection, represent a small fraction of T cells infused at transplantation.
Collapse
Affiliation(s)
- Marina Deschamps
- Institut National de la Santé et de la Recherche Médicale, U645, Besançon, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Developments in the treatment of sickle cell disease (SCD) have not kept pace with advances in understanding the pathophysiology of this haemoglobinopathy. Drugs undergoing preclinical and clinical assessment for the therapy of these globin gene disorders are discussed in this article. Beginning with investigational agents for treatment of SCD as a whole, the discussion proceeds to drugs being developed for specific manifestations or iatrogenic complications. Despite being licensed in the USA, the prototype antisickling agent, hydroxycarbamide, has not attained worldwide clinical use because of concerns about long-term toxicity. The less toxic decitabine, which (as with hydroxycarbamide) increases fetal haemoglobin level, cannot be administered orally; therefore, the search continues for effective and safe antisickling drugs that can be taken orally. The naturally occurring benzaldehyde 5-hydroxymethyl-2-furfural has shown promising antisickling properties in vitro, and when administered to transgenic sickle mice. These effects are surpassed by the new synthetic pyridyl derivatives of benzaldehyde. Studies in humans with SCD are required to assess the clinical efficacy of these benzaldehydes. Niprisan, another antisickling agent with significant clinical efficacy and an attractive safety profile, is undergoing further development. The prospects of antiadhesion therapy in SCD are demonstrated by a recombinant protein containing the Fc fragment of IgG fused to the natural ligand for selectins: the conjugate significantly inhibited blood vessel occlusion in transgenic sickle mice. Whereas the orally administrable iron-chelating agent deferasirox is likely to increasingly take the place of desferioxamine (which can only be given parenterally), effective treatment of priapism in SCD remains a distressing challenge.
Collapse
MESH Headings
- Acetamides/pharmacology
- Acetamides/therapeutic use
- Anemia, Sickle Cell/complications
- Anemia, Sickle Cell/drug therapy
- Anemia, Sickle Cell/metabolism
- Anemia, Sickle Cell/therapy
- Animals
- Antihypertensive Agents/therapeutic use
- Antisickling Agents/pharmacology
- Antisickling Agents/therapeutic use
- Benzaldehydes/pharmacology
- Benzaldehydes/therapeutic use
- Benzoates/administration & dosage
- Benzoates/therapeutic use
- Carnitine/therapeutic use
- Cell Adhesion
- Deferasirox
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Etilefrine/therapeutic use
- Female
- Genetic Therapy/methods
- Hematopoietic Stem Cell Transplantation
- Humans
- Hydroxyurea/pharmacology
- Hydroxyurea/therapeutic use
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/etiology
- Iron Chelating Agents/administration & dosage
- Iron Chelating Agents/therapeutic use
- Male
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/pharmacology
- Membrane Glycoproteins/therapeutic use
- Potassium Channels, Calcium-Activated/antagonists & inhibitors
- Potassium Channels, Calcium-Activated/metabolism
- Priapism/drug therapy
- Priapism/etiology
- Recombinant Fusion Proteins/pharmacology
- Recombinant Fusion Proteins/therapeutic use
- Triazoles/administration & dosage
- Triazoles/therapeutic use
- Trityl Compounds/pharmacology
- Trityl Compounds/therapeutic use
- Vasoconstrictor Agents/therapeutic use
Collapse
Affiliation(s)
- Iheanyi Okpala
- St Thomas' Hospital, University of London, London SE1 7EH, UK.
| |
Collapse
|
49
|
Breckpot K, Aerts JL, Thielemans K. Lentiviral vectors for cancer immunotherapy: transforming infectious particles into therapeutics. Gene Ther 2007; 14:847-62. [PMID: 17361214 DOI: 10.1038/sj.gt.3302947] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Lentiviral vectors have emerged as promising tools for both gene therapy and immunotherapy purposes. They exhibit several advantages over other viral systems in that they are less immunogenic and are capable of transducing a wide range of different cell types, including dendritic cells (DC). DC transduced ex vivo with a whole range of different (tumor) antigens were capable of inducing strong antigen-specific T-cell responses, both in vitro and in vivo. Recently, the administration of lentiviral vectors in vivo has gained substantial interest as an alternative method for antigen-specific immunization. This method offers a number of advantages over DC vaccines as the same lentivirus can in principle be used for all patients resulting in a significantly reduced cost and requirement for considerably less expertise for the generation and administration of lentiviral vaccines. By selectively targeting lentiviral vectors to, or restricting transgene expression in certain cell types, selectivity, safety and efficacy can be further improved. This review will focus on the use of direct administration of lentiviral vectors encoding tumor-associated antigens (TAA) for the induction of tumor-specific immune responses in vivo, with a special focus on problems related to the generation of large amounts of highly purified virus and specific targeting of antigen-presenting cells (APC).
Collapse
Affiliation(s)
- K Breckpot
- Laboratory of Molecular and Cellular Therapy, Department of Physiology and Immunology, Medical School of the Vrije Universiteit Brussel, Brussels, Belgium.
| | | | | |
Collapse
|
50
|
Mok HP, Javed S, Lever A. Stable gene expression occurs from a minority of integrated HIV-1-based vectors: transcriptional silencing is present in the majority. Gene Ther 2007; 14:741-51. [PMID: 17330088 DOI: 10.1038/sj.gt.3302923] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Human immunodeficiency virus (HIV)-based vectors are being increasingly used in vitro for gene transfer and in vivo for gene therapy. The proportion of integrated retroviral vectors that are silenced or remain transcriptionally active, and the stability of gene expression in the latter remains poorly explored. To study this, T cells were infected with an HIV-1-based vector construct containing a long terminal repeat-driven reporter gene. Only a small percentage of detectable integrated vector expressed gene product. In clones derived from cells with transcriptionally active vector, gene expression was remarkably stable with more than 80% continuing to express for greater than 18 months. Failure to continue expressing the vector was associated with epigenetic changes. Our data suggest that there are two forms of vector silencing: one occurring immediately after integration affecting the majority of the vectors, and one occurring in the much longer term affecting a small minority of vectors which had previously established expression.
Collapse
Affiliation(s)
- H P Mok
- Department of Medicine, University of Cambridge, Level 5, Addenbrooke's Hospital, Cambridge, UK
| | | | | |
Collapse
|