1
|
Adabi E, Charitidis FT, Thalheimer FB, Guaza-Lasheras M, Clarke C, Buchholz CJ. Enhanced conversion of T cells into CAR T cells by modulation of the MAPK/ERK pathway. Cell Rep Med 2025; 6:101970. [PMID: 39938523 PMCID: PMC11866553 DOI: 10.1016/j.xcrm.2025.101970] [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: 07/30/2024] [Revised: 11/20/2024] [Accepted: 01/22/2025] [Indexed: 02/14/2025]
Abstract
Delivery of chimeric antigen receptors (CARs) to T cells is usually mediated by lentiviral vectors (LVs), which can have broad tropism or be T cell targeted. To better understand the molecular events during CAR T cell generation, T cell transduction with four different LVs is followed by single-cell multi-omics analysis, distinguishing between transduced T cells and T cells with vector signal but no CAR. We find that only a fraction of the T cells that encounter vectors convert into CAR T cells. Single-cell transcriptome data reveal that interferon-stimulated genes are upregulated in non-transduced cells, whereas extracellular signal-regulated kinase (ERK)2 phosphatases are upregulated in CAR T cells. This expression pattern is evident in CAR T cells from healthy donors and patients. The role of the mitogen-activated protein kinase (MAPK)/ERK pathway in CAR T cell generation is confirmed by chemical inhibitors. These data provide molecular insights into T cell transduction with implications for improving CAR T cell generation.
Collapse
Affiliation(s)
- Elham Adabi
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Paul-Ehrlich-Strasse 51-59, 63225 Langen, Hessen, Germany
| | - Filippos T Charitidis
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Paul-Ehrlich-Strasse 51-59, 63225 Langen, Hessen, Germany
| | - Frederic B Thalheimer
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Paul-Ehrlich-Strasse 51-59, 63225 Langen, Hessen, Germany; Hematology, Cell and Gene Therapy, Paul-Ehrlich-Institut, Paul-Ehrlich-Strasse 51-59, 63225 Langen, Hessen, Germany; Frankfurt Cancer Institute (FCI), Goethe University, 60590 Frankfurt am Main, Germany
| | - Mar Guaza-Lasheras
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Paul-Ehrlich-Strasse 51-59, 63225 Langen, Hessen, Germany
| | - Colin Clarke
- National Institute for Bioprocessing Research and Training, Fosters Avenue, A94 X099 Blackrock, Dublin, Ireland; School of Chemical and Bioprocess Engineering, University College Dublin, D04 V1W8 Belfield, Dublin, Ireland
| | - Christian J Buchholz
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Paul-Ehrlich-Strasse 51-59, 63225 Langen, Hessen, Germany; Frankfurt Cancer Institute (FCI), Goethe University, 60590 Frankfurt am Main, Germany; Deutsches Krebsforschungszentrum and German Cancer Consortium (DKTK), 69120 Heidelberg, Germany.
| |
Collapse
|
2
|
Kontogiannis T, Braybrook J, McElroy C, Foy C, Whale AS, Quaglia M, Smales CM. Characterization of AAV vectors: A review of analytical techniques and critical quality attributes. Mol Ther Methods Clin Dev 2024; 32:101309. [PMID: 39234444 PMCID: PMC11372808 DOI: 10.1016/j.omtm.2024.101309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2024]
Abstract
Standardized evaluation of adeno-associated virus (AAV) vector products for biotherapeutic application is essential to ensure the safety and efficacy of gene therapies. This includes analyzing the critical quality attributes of the product. However, many of the current analytical techniques used to assess these attributes have limitations, including low throughput, large sample requirements, poorly understood measurement variability, and lack of comparability between methods. To address these challenges, it is essential to establish higher-order reference methods that can be used for comparability measurements, optimization of current assays, and development of reference materials. Highly precise methods are necessary for measuring the empty/partial/full capsid ratios and the titer of AAV vectors. Additionally, it is important to develop methods for the measurement of less-established critical quality attributes, including post-translational modifications, capsid stoichiometry, and methylation profiles. By doing so, we can gain a better understanding of the influence of these attributes on the quality of the product. Moreover, quantification of impurities, such as host-cell proteins and DNA contaminants, is crucial for obtaining regulatory approval. The development and application of refined methodologies will be essential to thoroughly characterize AAV vectors by informing process development and facilitating the generation of reference materials for assay validation and calibration.
Collapse
Affiliation(s)
- Theodoros Kontogiannis
- School of Biosciences, Division of Natural Sciences, University of Kent, Canterbury, Kent CT2 7NJ, UK
- National Measurement Laboratory at LGC, Teddington, Middlesex TW11 0LY, UK
| | - Julian Braybrook
- National Measurement Laboratory at LGC, Teddington, Middlesex TW11 0LY, UK
| | | | - Carole Foy
- National Measurement Laboratory at LGC, Teddington, Middlesex TW11 0LY, UK
| | - Alexandra S Whale
- National Measurement Laboratory at LGC, Teddington, Middlesex TW11 0LY, UK
| | - Milena Quaglia
- Reading Scientific Services Ltd, Reading Science Centre, Whiteknights Campus, Pepper Lane, Reading Berkshire RG6 6LA, UK
| | - C Mark Smales
- School of Biosciences, Division of Natural Sciences, University of Kent, Canterbury, Kent CT2 7NJ, UK
- National Institute for Bioprocessing Research and Training, Blackrock, Co, Foster Avenue, A94 X099 Mount Merrion, Dublin, Ireland
| |
Collapse
|
3
|
Ingusci S, Hall BL, Goins WF, Cohen JB, Glorioso JC. Viral vectors for gene delivery to the central nervous system. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:59-81. [PMID: 39341663 DOI: 10.1016/b978-0-323-90120-8.00001-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Brain diseases with a known or suspected genetic basis represent an important frontier for advanced therapeutics. The central nervous system (CNS) is an intricate network in which diverse cell types with multiple functions communicate via complex signaling pathways, making therapeutic intervention in brain-related diseases challenging. Nevertheless, as more information on the molecular genetics of brain-related diseases becomes available, genetic intervention using gene therapeutic strategies should become more feasible. There remain, however, several significant hurdles to overcome that relate to (i) the development of appropriate gene vectors and (ii) methods to achieve local or broad vector delivery. Clearly, gene delivery tools must be engineered for distribution to the correct cell type in a specific brain region and to accomplish therapeutic transgene expression at an appropriate level and duration. They also must avoid all toxicity, including the induction of inflammatory responses. Over the last 40 years, various types of viral vectors have been developed as tools to introduce therapeutic genes into the brain, primarily targeting neurons. This review describes the most prominent vector systems currently approaching clinical application for CNS disorders and highlights both remaining challenges as well as improvements in vector designs that achieve greater safety, defined tropism, and therapeutic gene expression.
Collapse
Affiliation(s)
- Selene Ingusci
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Bonnie L Hall
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - William F Goins
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Justus B Cohen
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Joseph C Glorioso
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
4
|
Charitidis FT, Adabi E, Ho N, Braun AH, Tierney C, Strasser L, Thalheimer FB, Childs L, Bones J, Clarke C, Buchholz CJ. CAR Gene Delivery by T-cell Targeted Lentiviral Vectors is Enhanced by Rapamycin Induced Reduction of Antiviral Mechanisms. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302992. [PMID: 37904721 PMCID: PMC10724389 DOI: 10.1002/advs.202302992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 09/11/2023] [Indexed: 11/01/2023]
Abstract
Lentiviral vectors (LV) have become the dominant tool for stable gene transfer into lymphocytes including chimeric antigen receptor (CAR) gene delivery to T cells, a major breakthrough in cancer therapy. Yet, room for improvement remains, especially for the latest LV generations delivering genes selectively into T cell subtypes, a key requirement for in vivo CAR T cell generation. Toward improving gene transfer rates with these vectors, whole transcriptome analyses on human T lymphocytes are conducted after exposure to CAR-encoding conventional vectors (VSV-LV) and vectors targeted to CD8+ (CD8-LV) or CD4+ T cells (CD4-LV). Genes related to quiescence and antiviral restriction are found to be upregulated in CAR-negative cells exposed to all types of LVs. Down-modulation of various antiviral restriction factors, including the interferon-induced transmembrane proteins (IFITMs) is achieved with rapamycin as verified by mass spectrometry (LC-MS). Strikingly, rapamycin enhances transduction by up to 7-fold for CD8-LV and CD4-LV without compromising CAR T cell activities but does not improve VSV-LV. When administered to humanized mice, CD8-LV results in higher rates of green fluorescent protein (GFP) gene delivery. Also in vivo CAR T cell generation is improved in kinetics and tumor control, however to a moderate extent, leaving room for improvement by optimizing the rapamycin administration schedule. The data favor multi-omics approaches for improvements in gene delivery.
Collapse
Affiliation(s)
| | - Elham Adabi
- Molecular Biotechnology and Gene TherapyPaul‐Ehrlich‐Institut63225LangenGermany
| | - Naphang Ho
- Molecular Biotechnology and Gene TherapyPaul‐Ehrlich‐Institut63225LangenGermany
| | - Angela H Braun
- Molecular Biotechnology and Gene TherapyPaul‐Ehrlich‐Institut63225LangenGermany
- Deutsches Krebsforschungszentrum and German Cancer Consortium (DKTK)69120HeidelbergGermany
| | - Ciara Tierney
- Characterisation and Comparability LaboratoryNational Institute for Bioprocessing Research and TrainingFoster Avenue, Mount Merrion, BlackrockDublinA94 X099Ireland
| | - Lisa Strasser
- Characterisation and Comparability LaboratoryNational Institute for Bioprocessing Research and TrainingFoster Avenue, Mount Merrion, BlackrockDublinA94 X099Ireland
| | - Frederic B Thalheimer
- Molecular Biotechnology and Gene TherapyPaul‐Ehrlich‐Institut63225LangenGermany
- Frankfurt Cancer Institute (FCI)Goethe University60590Frankfurt am MainGermany
| | - Liam Childs
- Host‐Pathogen InteractionsPaul‐Ehrlich‐Institut63225LangenGermany
| | - Jonathan Bones
- Characterisation and Comparability LaboratoryNational Institute for Bioprocessing Research and TrainingFoster Avenue, Mount Merrion, BlackrockDublinA94 X099Ireland
- School of Chemical and Bioprocess EngineeringUniversity College DublinD04 V1W8BelfieldDublinIreland
| | - Colin Clarke
- Characterisation and Comparability LaboratoryNational Institute for Bioprocessing Research and TrainingFoster Avenue, Mount Merrion, BlackrockDublinA94 X099Ireland
- National Institute for Bioprocessing Research and TrainingA94×099Foster Avenue, Mount Merrion, BlackrockDublinIreland
| | - Christian J Buchholz
- Molecular Biotechnology and Gene TherapyPaul‐Ehrlich‐Institut63225LangenGermany
- Deutsches Krebsforschungszentrum and German Cancer Consortium (DKTK)69120HeidelbergGermany
- Frankfurt Cancer Institute (FCI)Goethe University60590Frankfurt am MainGermany
| |
Collapse
|
5
|
Song Y, Wang N, Shi H, Zhang D, Wang Q, Guo S, Yang S, Ma J. Biomaterials combined with ADSCs for bone tissue engineering: current advances and applications. Regen Biomater 2023; 10:rbad083. [PMID: 37808955 PMCID: PMC10551240 DOI: 10.1093/rb/rbad083] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 08/07/2023] [Accepted: 08/31/2023] [Indexed: 10/10/2023] Open
Abstract
In recent decades, bone tissue engineering, which is supported by scaffold, seed cells and bioactive molecules (BMs), has provided new hope and direction for treating bone defects. In terms of seed cells, compared to bone marrow mesenchymal stem cells, which were widely utilized in previous years, adipose-derived stem cells (ADSCs) are becoming increasingly favored by researchers due to their abundant sources, easy availability and multi-differentiation potentials. However, there is no systematic theoretical basis for selecting appropriate biomaterials loaded with ADSCs. In this review, the regulatory effects of various biomaterials on the behavior of ADSCs are summarized from four perspectives, including biocompatibility, inflammation regulation, angiogenesis and osteogenesis, to illustrate the potential of combining various materials with ADSCs for the treatment of bone defects. In addition, we conclude the influence of additional application of various BMs on the bone repair effect of ADSCs, in order to provide more evidences and support for the selection or preparation of suitable biomaterials and BMs to work with ADSCs. More importantly, the associated clinical case reports and experiments are generalized to provide additional ideas for the clinical transformation and application of bone tissue engineering loaded with ADSCs.
Collapse
Affiliation(s)
- Yiping Song
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Ning Wang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Huixin Shi
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Dan Zhang
- School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
- Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang 110001, China
| | - Qiang Wang
- School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
- Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang 110001, China
| | - Shu Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Shude Yang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
- Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang 110001, China
| | - Jia Ma
- School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
- Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang 110001, China
| |
Collapse
|
6
|
Host Cell Restriction Factors Blocking Efficient Vector Transduction: Challenges in Lentiviral and Adeno-Associated Vector Based Gene Therapies. Cells 2023; 12:cells12050732. [PMID: 36899868 PMCID: PMC10001033 DOI: 10.3390/cells12050732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/02/2023] [Accepted: 02/08/2023] [Indexed: 03/03/2023] Open
Abstract
Gene therapy relies on the delivery of genetic material to the patient's cells in order to provide a therapeutic treatment. Two of the currently most used and efficient delivery systems are the lentiviral (LV) and adeno-associated virus (AAV) vectors. Gene therapy vectors must successfully attach, enter uncoated, and escape host restriction factors (RFs), before reaching the nucleus and effectively deliver the therapeutic genetic instructions to the cell. Some of these RFs are ubiquitously expressed in mammalian cells, while others are cell-specific, and others still are expressed only upon induction by danger signals as type I interferons. Cell restriction factors have evolved to protect the organism against infectious diseases and tissue damage. These restriction factors can be intrinsic, directly acting on the vector, or related with the innate immune response system, acting indirectly through the induction of interferons, but both are intertwined. The innate immunity is the first line of defense against pathogens and, as such cells derived from myeloid progenitors (but not only), are well equipped with RFs to detect pathogen-associated molecular patterns (PAMPs). In addition, some non-professional cells, such as epithelial cells, endothelial cells, and fibroblasts, play major roles in pathogen recognition. Unsurprisingly, foreign DNA and RNA molecules are among the most detected PAMPs. Here, we review and discuss identified RFs that block LV and AAV vector transduction, hindering their therapeutic efficacy.
Collapse
|
7
|
Sertkaya H, Hidalgo L, Ficarelli M, Kmiec D, Signell AW, Ali S, Parker H, Wilson H, Neil SJ, Malim MH, Vink CA, Swanson CM. Minimal impact of ZAP on lentiviral vector production and transduction efficiency. Mol Ther Methods Clin Dev 2021; 23:147-157. [PMID: 34703838 PMCID: PMC8517000 DOI: 10.1016/j.omtm.2021.08.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 08/24/2021] [Indexed: 11/29/2022]
Abstract
The antiviral protein ZAP binds CpG dinucleotides in viral RNA to inhibit replication. This has likely led to the CpG suppression observed in many RNA viruses, including retroviruses. Sequences added to retroviral vector genomes, such as internal promoters, transgenes, or regulatory elements, substantially increase CpG abundance. Because these CpGs could allow retroviral vector RNA to be targeted by ZAP, we analyzed whether it restricts vector production, transduction efficiency, and transgene expression. Surprisingly, even though CpG-high HIV-1 was efficiently inhibited by ZAP in HEK293T cells, depleting ZAP did not substantially increase lentiviral vector titer using several packaging and genome plasmids. ZAP overexpression also did not inhibit lentiviral vector titer. In addition, decreasing CpG abundance in a lentiviral vector genome did not increase its titer, and a gammaretroviral vector derived from murine leukemia virus was not substantially restricted by ZAP. Overall, we show that the increased CpG abundance in retroviral vectors relative to the wild-type retroviruses they are derived from does not intrinsically sensitize them to ZAP. Further understanding of how ZAP specifically targets transcripts to inhibit their expression may allow the development of CpG sequence contexts that efficiently recruit or evade this antiviral system.
Collapse
Affiliation(s)
- Helin Sertkaya
- Department of Infectious Diseases, King’s College London, London SE1 9RT, UK
| | - Laura Hidalgo
- Department of Infectious Diseases, King’s College London, London SE1 9RT, UK
| | - Mattia Ficarelli
- Department of Infectious Diseases, King’s College London, London SE1 9RT, UK
| | - Dorota Kmiec
- Department of Infectious Diseases, King’s College London, London SE1 9RT, UK
| | - Adrian W. Signell
- Department of Infectious Diseases, King’s College London, London SE1 9RT, UK
| | - Sadfer Ali
- Cell & Gene Therapy Platform, Medicinal Science and Technology, GSK, Stevenage SG1 2NY, UK
| | - Hannah Parker
- Department of Infectious Diseases, King’s College London, London SE1 9RT, UK
| | - Harry Wilson
- Department of Infectious Diseases, King’s College London, London SE1 9RT, UK
| | - Stuart J.D. Neil
- Department of Infectious Diseases, King’s College London, London SE1 9RT, UK
| | - Michael H. Malim
- Department of Infectious Diseases, King’s College London, London SE1 9RT, UK
| | - Conrad A. Vink
- Cell & Gene Therapy Platform, Medicinal Science and Technology, GSK, Stevenage SG1 2NY, UK
| | - Chad M. Swanson
- Department of Infectious Diseases, King’s College London, London SE1 9RT, UK
| |
Collapse
|
8
|
Genetic in vivo engineering of human T lymphocytes in mouse models. Nat Protoc 2021; 16:3210-3240. [PMID: 33846629 DOI: 10.1038/s41596-021-00510-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 01/21/2021] [Indexed: 12/19/2022]
Abstract
Receptor targeting of vector particles is a key technology to enable cell type-specific in vivo gene delivery. For example, T cells in humanized mouse models can be modified by lentiviral vectors (LVs) targeted to human T-cell markers to enable them to express chimeric antigen receptors (CARs). Here, we provide detailed protocols for the generation of CD4- and CD8-targeted LVs (which takes ~9 d in total). We also describe how to humanize immunodeficient mice with hematopoietic stem cells (which takes 12-16 weeks) and precondition (over 5 d) and administer the vector stocks. Conversion of the targeted cell type is monitored by PCR and flow cytometry of blood samples. A few weeks after administration, ~10% of the targeted T-cell subtype can be expected to have converted to CAR T cells. By closely following the protocol, sufficient vector stock for the genetic manipulation of 10-15 humanized mice is obtained. We also discuss how the protocol can be easily adapted to use LVs targeted to other types of receptors and/or for delivery of other genes of interest.
Collapse
|
9
|
Sauter MM, Brandt CR. Knockdown of TRIM5α or TRIM11 increases lentiviral vector transduction efficiency of human Muller cells. Exp Eye Res 2021; 204:108436. [PMID: 33440192 DOI: 10.1016/j.exer.2021.108436] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/17/2020] [Accepted: 01/02/2021] [Indexed: 02/07/2023]
Abstract
The goal of this study was to determine the expression and distribution of the host restriction factors (RFs) TRIM5α and TRIM11 in non-human primate (NHP) neural retina tissue and the human Muller cell line MIO-M1. In addition, experiments were performed to determine the effect of TRIM5α and TRIM11 knockdown on FIVGFP transduction of MIO-M1 cells with the goal of devising strategies to increase the efficiency of lentiviral (LV) gene delivery. Immunofluorescence (IF) studies indicated that TRIM5α and TRIM11 were localized predominantly in nuclei within the outer nuclear layer (ONL) and inner nuclear layer (INL) of NHP retina tissue. Double label IF indicated that TRIM5α and TRIM11 were localized to some of the retinal Muller cell nuclei. MIO-M1 cells expressed TRIM5α predominantly in the nucleus and TRIM11 primarily in the cytosol. FIVGFP transduction efficiency was significantly increased, at 4 and 7 days post transduction, in TRIM5α and TRIM11 knockdown clones (KD) compared to WT MIO-M1 cells. In addition, pretreatment with the proteasome inhibitor MG132 increased the transduction efficiency of FIVGFP in WT MIO-M1 cells. The nuclear translocation of NF-κB (p65), at 72 h post FIVGFP transduction, was enhanced in TRIM5α and TRIM11 KD clones. The expression of TRIM5α and TRIM11 in macaque neural retina tissue and MIO-M1 cells indicate the presence of these RFs in NHP retina and human Muller cells. Our data indicate that even partial knockdown of TRIM5α or TRIM11, or a short proteasome inhibitor pretreatment, can increase the transduction efficiency of a LV vector.
Collapse
Affiliation(s)
- Monica M Sauter
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Curtis R Brandt
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53706, USA; Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, 53706, USA; McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, 53706, USA.
| |
Collapse
|
10
|
Abstract
Several viral vector-based gene therapy drugs have now received marketing approval. A much larger number of additional viral vectors are in various stages of clinical trials for the treatment of genetic and acquired diseases, with many more in pre-clinical testing. Efficiency of gene transfer and ability to provide long-term therapy make these vector systems very attractive. In fact, viral vector gene therapy has been able to treat or even cure diseases for which there had been no or only suboptimal treatments. However, innate and adaptive immune responses to these vectors and their transgene products constitute substantial hurdles to clinical development and wider use in patients. This review provides an overview of the type of immune responses that have been documented in animal models and in humans who received gene transfer with one of three widely tested vector systems, namely adenoviral, lentiviral, or adeno-associated viral vectors. Particular emphasis is given to mechanisms leading to immune responses, efforts to reduce vector immunogenicity, and potential solutions to the problems. At the same time, we point out gaps in our knowledge that should to be filled and problems that need to be addressed going forward.
Collapse
Affiliation(s)
- Jamie L Shirley
- Gene Therapy Center, University of Massachusetts, Worchester, MA, USA
| | - Ype P de Jong
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, NY, USA
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, MA, USA
| | - Roland W Herzog
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
11
|
Rodriguez M, Porada CD, Almeida-Porada G. Mechanistic Insights into Factor VIII Immune Tolerance Induction via Prenatal Cell Therapy in Hemophilia A. CURRENT STEM CELL REPORTS 2019; 5:145-161. [PMID: 32351874 DOI: 10.1007/s40778-019-00165-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Purpose of Review Prenatal stem cell and gene therapy approaches are amongst the few therapies that can promise the birth of a healthy infant with specific known genetic diseases. This review describes fetal immune cell signaling and its potential influence on donor cell engraftment, and summarizes mechanisms of central T cell tolerance to peripherally-acquired antigen in the context of prenatal therapies for Hemophilia A. Recent Findings During early gestation, different subsets of antigen presenting cells take up peripherally-acquired, non-inherited antigens and induce the deletion of antigen-reactive T-cell precursors in the thymus, demonstrating the potential for using prenatal cell and gene therapies to induce central tolerance to FVIII in the context of prenatal diagnosis/therapy of Hemophilia A. Summary Prenatal cell and gene therapies are promising approaches to treat several genetic disorders including Hemophilia A and B. Understanding the mechanisms of how FVIII-specific tolerance is achieved during ontogeny could help develop novel therapies for HA and better approaches to overcome FVIII inhibitors.
Collapse
Affiliation(s)
- Martin Rodriguez
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Christopher D Porada
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Graҫa Almeida-Porada
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
12
|
Maes ME, Colombo G, Schulz R, Siegert S. Targeting microglia with lentivirus and AAV: Recent advances and remaining challenges. Neurosci Lett 2019; 707:134310. [PMID: 31158432 PMCID: PMC6734419 DOI: 10.1016/j.neulet.2019.134310] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 05/30/2019] [Indexed: 02/06/2023]
Abstract
Microglia have emerged as a critical component of neurodegenerative diseases. Genetic manipulation of microglia can elucidate their functional impact in disease. In neuroscience, recombinant viruses such as lentiviruses and adeno-associated viruses (AAVs) have been successfully used to target various cell types in the brain, although effective transduction of microglia is rare. In this review, we provide a short background of lentiviruses and AAVs, and strategies for designing recombinant viral vectors. Then, we will summarize recent literature on successful microglial transductions in vitro and in vivo, and discuss the current challenges. Finally, we provide guidelines for reporting the efficiency and specificity of viral targeting in microglia, which will enable the microglial research community to assess and improve methodologies for future studies.
Collapse
Affiliation(s)
- Margaret E Maes
- Institute of Science and Technology (IST) Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Gloria Colombo
- Institute of Science and Technology (IST) Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Rouven Schulz
- Institute of Science and Technology (IST) Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Sandra Siegert
- Institute of Science and Technology (IST) Austria, Am Campus 1, 3400 Klosterneuburg, Austria.
| |
Collapse
|
13
|
Kim I, Lee SS, Kim SHL, Bae S, Lee H, Hwang NS. Osteogenic Effects of VEGF-Overexpressed Human Adipose-Derived Stem Cells with Whitlockite Reinforced Cryogel for Bone Regeneration. Macromol Biosci 2019; 19:e1800460. [PMID: 30821921 DOI: 10.1002/mabi.201800460] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 02/13/2019] [Indexed: 12/13/2022]
Abstract
Bone is a vascularized tissue that is comprised of collagen fibers and calcium phosphate crystals such as hydroxyapatite (HAp) and whitlockite (WH). HAp and WH are known to elicit bone regeneration by stimulating osteoblast activities and osteogenic commitment of stem cells. In addition, vascular endothelial growth factor (VEGF) is shown to promote osteogenesis and angiogenesis which is considered as an essential process in bone repair by providing nutrients. In this study, VEGF-secreting human adipose-derived stem cells (VEGF-ADSCs) are developed by transducing ADSCs with VEGF-encoded lentivirus. Additionally, WH-reinforced gelatin/heparin cryogels (WH-C) are fabricated by loading WH into gelatin/heparin cryogels. VEGF-ADSC secrete tenfold more VEGF than ADSC and show increased VEGF secretion with cell growth. Also, incorporation of WH into cryogels provides a mineralized environment with ions secreted from WH. When the VEGF-ADSCs are seeded on WH-C, sustained release of VEGF is observed due to the specific affinity of VEGF to heparin. Finally, the synergistic effect of VEGF-ADSC and WH on osteogenesis is successfully confirmed by alkaline phosphatase and real-time polymerase chain reaction analysis. In vivo bone formation is demonstrated via implantation of VEGF-ADSC seeded WH-C into mouse calvarial bone defect model, resulted in enhanced bone development with the highest bone volume/total volume.
Collapse
Affiliation(s)
- Inseon Kim
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seunghun S Lee
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seung Hyun L Kim
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sunghoon Bae
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hoyon Lee
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Nathaniel S Hwang
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea.,Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea.,BioMax Institute of Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
14
|
Alfranca A, Campanero MR, Redondo JM. New Methods for Disease Modeling Using Lentiviral Vectors. Trends Mol Med 2018; 24:825-837. [PMID: 30213701 DOI: 10.1016/j.molmed.2018.08.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/02/2018] [Accepted: 08/03/2018] [Indexed: 12/11/2022]
Abstract
Lentiviral vectors (LVs) transduce quiescent cells and provide stable integration to maintain transgene expression. Several approaches have been adopted to optimize LV safety profiles. Similarly, LV targeting has been tailored through strategies including the modification of envelope components, the use of specific regulatory elements, and the selection of appropriate administration routes. Models of aortic disease based on a single injection of pleiotropic LVs have been developed that efficiently transduce the three aorta layers in wild type mice. This approach allows the dissection of pathways involved in aortic aneurysm formation and the identification of targets for gene therapy in aortic diseases. LVs provide a fast, efficient, and affordable alternative to genetically modified mice to study disease mechanisms and develop therapeutic tools.
Collapse
Affiliation(s)
- Arantzazu Alfranca
- Department of Immunology, Hospital Universitario de La Princesa, Madrid, Spain; CIBERCV, Madrid, Spain.
| | - Miguel R Campanero
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid, Spain; CIBERCV, Madrid, Spain
| | - Juan Miguel Redondo
- Gene Regulation in Cardiovascular Remodeling and Inflammation Group, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain; CIBERCV, Madrid, Spain.
| |
Collapse
|
15
|
Peng W, Zhang J, Zhang H, Liu G, Dong W, Zhang F. Effects of lentiviral transfection containing bFGF gene on the biological characteristics of rabbit BMSCs. J Cell Biochem 2018; 119:8389-8397. [DOI: 10.1002/jcb.27034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 04/06/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Wuxun Peng
- Department of Emergency OrthopedicsAffiliated Hospital of Guizhou Medical UniversityChina
| | - Jian Zhang
- Department of Emergency OrthopedicsAffiliated Hospital of Guizhou Medical UniversityChina
| | - Huai Zhang
- Department of Emergency OrthopedicsAffiliated Hospital of Guizhou Medical UniversityChina
| | - Gang Liu
- Department of Emergency OrthopedicsAffiliated Hospital of Guizhou Medical UniversityChina
| | - Wentao Dong
- Department of Emergency OrthopedicsAffiliated Hospital of Guizhou Medical UniversityChina
| | - Fei Zhang
- Department of Emergency OrthopedicsAffiliated Hospital of Guizhou Medical UniversityChina
| |
Collapse
|
16
|
Gunasekaran V, Selvaraj R. Biological Pacemakers – A Review. INTERNATIONAL JOURNAL OF CARDIOVASCULAR PRACTICE 2018. [DOI: 10.21859/ijcp-03103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
17
|
Suwanmanee T, Ferris MT, Hu P, Gui T, Montgomery SA, Pardo-Manuel de Villena F, Kafri T. Toward Personalized Gene Therapy: Characterizing the Host Genetic Control of Lentiviral-Vector-Mediated Hepatic Gene Delivery. Mol Ther Methods Clin Dev 2017; 5:83-92. [PMID: 28480308 PMCID: PMC5415322 DOI: 10.1016/j.omtm.2017.03.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 03/30/2017] [Indexed: 12/21/2022]
Abstract
The success of lentiviral vectors in curing fatal genetic and acquired diseases has opened a new era in human gene therapy. However, variability in the efficacy and safety of this therapeutic approach has been reported in human patients. Consequently, lentiviral-vector-based gene therapy is limited to incurable human diseases, with little understanding of the underlying causes of adverse effects and poor efficacy. To assess the role that host genetic variation has on efficacy of gene therapy, we characterized lentiviral-vector gene therapy within a set of 12 collaborative cross mouse strains. Lentiviral vectors carrying the firefly luciferase cDNA under the control of a liver-specific promoter were administered to female mice, with total-body and hepatic luciferase expression periodically monitored through 41 weeks post-vector administration. Vector copy number per diploid genome in mouse liver and spleen was determined at the end of this study. We identified major strain-specific contributions to overall success of transduction, vector biodistribution, maximum luciferase expression, and the kinetics of luciferase expression throughout the study. Our results highlight the importance of genetic variation on gene-therapeutic efficacy; provide new models with which to more rigorously assess gene therapy approaches; and suggest that redesigning preclinical studies of gene-therapy methodologies might be appropriate.
Collapse
Affiliation(s)
- Thipparat Suwanmanee
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Martin T. Ferris
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Peirong Hu
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tong Gui
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Stephanie A. Montgomery
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Fernando Pardo-Manuel de Villena
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tal Kafri
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|