1
|
Sheehan M, Kumpf SW, Qian J, Rubitski DM, Oziolor E, Lanz TA. Comparison and cross-validation of long-read and short-read target-enrichment sequencing methods to assess AAV vector integration into host genome. Mol Ther Methods Clin Dev 2024; 32:101352. [PMID: 39506980 PMCID: PMC11539135 DOI: 10.1016/j.omtm.2024.101352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024]
Abstract
Evaluation of host integration profiles by adeno-associated virus (AAV) is an important component of de-risking novel AAV gene therapies. Targeted enrichment sequencing (TES) is a cost-effective and comprehensive method for assessing integration. Most published TES datasets have been generated using short-read sequencing, which enables quantitation of integration sites (ISs) and identifies patterns such as hotspots or clonal expansion. Characteristics such as IS length and recombination require longer reads to measure. The present study compared short-read to long-read TES using samples from monkeys treated with AAV and used in vitro lentiviral-treated samples, a stable cell line, and an engineered spike-in as controls. Both methods showed stochastic integration by both AAV and lentivirus, with most vector domains identified in ISs. More ISs were identified by short-read TES, as deeper coverage per base was achieved from a single sequencing run. AAV-treated samples showed minimal evidence of clonal expansion, in contrast to in vitro treated and stably transduced lentiviral cell line samples. Long-read TES revealed vector rearrangement in 4%-40% of ISs in AAV-treated animals. In summary, both methods yielded similar conclusions about relative numbers of ISs and overall patterns. Long-read TES identified fewer ISs but enabled measurement of IS length and recombination patterns.
Collapse
Affiliation(s)
- Mark Sheehan
- Global Discovery, Investigative and Translational Sciences, Pfizer Inc, Groton, CT 06340, USA
| | - Steven W. Kumpf
- Global Discovery, Investigative and Translational Sciences, Pfizer Inc, Groton, CT 06340, USA
| | - Jessie Qian
- Global Discovery, Investigative and Translational Sciences, Pfizer Inc, Groton, CT 06340, USA
| | - David M. Rubitski
- Global Discovery, Investigative and Translational Sciences, Pfizer Inc, Groton, CT 06340, USA
| | - Elias Oziolor
- Computational Safety Sciences, Pfizer Inc, Groton, CT 06340, USA
| | - Thomas A. Lanz
- Global Discovery, Investigative and Translational Sciences, Pfizer Inc, Groton, CT 06340, USA
| |
Collapse
|
2
|
Şeker ME, Erol ÖD, Pervin B, Wagemaker G, van Til NP, Aerts-Kaya F. Assessment of non-myelotoxic agents as a preparatory regimen for hematopoietic stem cell gene therapy. Hum Cell 2024; 38:9. [PMID: 39460845 DOI: 10.1007/s13577-024-01130-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024]
Abstract
RAG2 deficiency is characterized by a lack of B and T lymphocytes, causing severe lethal infections. Currently, RAG2 deficiency is treated with a Hematopoietic Stem Cell transplantation (HSCT). Most conditioning regimens used before HSCT consist of alkylating myelotoxic agents with or without irradiation and affect growth and development of pediatric patients. Here, we developed a non-myelotoxic regimen using G-CSF, VLA-4I or AMD3100. These agents are known HSC mobilizers or affect bone marrow (BM) permeability and may support the homing of HSCs to the BM, without inducing major side effects. Female Rag2-/- mice were pre-treated with Busulfan (BU), G-CSF, VLA-4I or AMD3100 and transplanted with male BM cells transduced with a lentiviral vector carrying codon optimized human RAG2 (RAG2co). Peripheral blood cell counts increased significantly after G-CSF, VLA-4I and AMD3100 treatment, but not after BU. Reconstitution of PB lymphocytes was comparable for all groups with full immune reconstitution at 6 months post transplantation, despite different methods of conditioning. Survival of mice pre-treated with non-myelotoxic agents was significantly higher than after BU treatment. Here, we show that the non-myelotoxic agents G-CSF, VLA-4I, and AMD3100 are highly effective as conditioning regimen before HSC gene therapy and can be used as an alternative to BU.
Collapse
Affiliation(s)
- Mehmet Emin Şeker
- Department of Stem Cell Sciences, Graduate School of Health Sciences, Hacettepe University, Ankara, Turkey
- Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey
| | - Özgür Doğuş Erol
- Department of Stem Cell Sciences, Graduate School of Health Sciences, Hacettepe University, Ankara, Turkey
- Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey
| | - Burcu Pervin
- Department of Stem Cell Sciences, Graduate School of Health Sciences, Hacettepe University, Ankara, Turkey
- Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey
| | - Gerard Wagemaker
- Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey
- Department of Hematology, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Niek P van Til
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Center, Amsterdam Neuroscience, 1081 HV, Amsterdam, The Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit, 1081 HV, Amsterdam, The Netherlands
| | - Fatima Aerts-Kaya
- Department of Stem Cell Sciences, Graduate School of Health Sciences, Hacettepe University, Ankara, Turkey.
- Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey.
- Hacettepe University Experimental Animals Application and Research Center (HÜDHAM), Hacettepe University, Ankara, Turkey.
- Hacettepe University Advanced Techologies Application and Research Center (HÜNİTEK), Hacettepe University, Ankara, Turkey.
| |
Collapse
|
3
|
Chan CH, Pearce DA. Demystifying gene(tic) therapies. Eur J Med Genet 2024; 71:104963. [PMID: 39069254 DOI: 10.1016/j.ejmg.2024.104963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/24/2024] [Accepted: 07/25/2024] [Indexed: 07/30/2024]
Abstract
This article summarizes the discussion from a session entitled "Demystifying gene therapies" that was held at the joint RE(ACT) congress and IRDiRC conference, 14-15 March 2023 in Berlin, Germany. The focus of this session was to discuss the changing landscape of genetic therapies and whether current resources exist to provide adequate education to stakeholders, such as researchers, clinicians, patient advocates, legislators, as well as the patients and their families. The goal of this article is not to provide a comprehensive overview of the current landscape in genetic therapies, but rather to highlight resources that may be useful to help "demystify" the myriad of genetic therapeutic approaches.
Collapse
Affiliation(s)
| | - David A Pearce
- Sanford Research, Sioux Falls, SD, USA; Sanford Health, Sioux Falls, SD, USA; Dept. of Pediatrics, Sanford School of Medicine, University of South Dakota, USA.
| |
Collapse
|
4
|
Klapwijk JC, Del Rio Espinola A, Libertini S, Collin P, Fellows MD, Jobling S, Lynch AM, Martus H, Vickers C, Zeller A, Biasco L, Brugman MH, Bushmann FD, Cathomen T, Ertl HCJ, Gabriel R, Gao G, Jadlowsky JK, Kimber I, Lanz TA, Levine BL, Micklethwaite KP, Onodera M, Pizzurro DM, Reed S, Rothe M, Sabatino DE, Salk JJ, Schambach A, Themis M, Yuan J. Improving the Assessment of Risk Factors Relevant to Potential Carcinogenicity of Gene Therapies: A Consensus Article. Hum Gene Ther 2024; 35:527-542. [PMID: 39049734 DOI: 10.1089/hum.2024.033] [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] [Indexed: 07/27/2024] Open
Abstract
Regulators and industry are actively seeking improvements and alternatives to current models and approaches to evaluate potential carcinogenicity of gene therapies (GTs). A meeting of invited experts was organized by NC3Rs/UKEMS (London, March 2023) to discuss this topic. This article describes the consensus reached among delegates on the definition of vector genotoxicity, sources of uncertainty, suitable toxicological endpoints for genotoxic assessment of GTs, and future research needs. The collected recommendations should inform the further development of regulatory guidelines for the nonclinical toxicological assessment of GT products.
Collapse
Affiliation(s)
| | | | | | - Philippe Collin
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Mick D Fellows
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Susan Jobling
- TestaVec Ltd, Maidenhead, United Kingdom
- Division of Biosciences, Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | | | | | - Catherine Vickers
- National Centre for the Replacement Refinement and Reduction of Animals in Research, London, United Kingdom
| | - Andreas Zeller
- F. Hoffmann-La Roche Ltd., pRED, Pharma Research & Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Luca Biasco
- UCL Zayed Centre for Research (ZCR), London, United Kingdom
| | - Martijn H Brugman
- Cell and Gene Therapy, GSK Medicine Research Centre, Stevenage, United Kingdom
| | - Frederic D Bushmann
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, Pennsylvania, USA
| | - Toni Cathomen
- Institute for Transfusion Medicine and Gene Therapy, Medical Center- University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hildegrund C J Ertl
- Ertl Laboratory, Vaccine & Immunotherapy Center, The Wistar Institute, Philadelphia, Pennsylvania, USA
| | | | - Guangping Gao
- Horae Gene Therapy Center, UMass Chan Medical School, University of Massachusetts, Worcester, Massachusetts, USA
| | - Julie K Jadlowsky
- Center for Cellular Immunotherapies and Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ian Kimber
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Thomas A Lanz
- Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut, USA
| | - Bruce L Levine
- Center for Cellular Immunotherapies and Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kenneth P Micklethwaite
- Department of Haematology, Blood Transplant and Cell Therapies Program, Westmead Hospital, Sydney, Australia
- NSW Health Pathology Blood Transplant and Cell Therapies Laboratory - ICPMR Westmead, Sydney, Australia
- Westmead Institute for Medical Research, Sydney, Australia
- Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Masafumi Onodera
- Gene & Cell Therapy Promotion Center, National Center for Child Health and Development, Tokyo, Japan
| | | | - Simon Reed
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Michael Rothe
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Denise E Sabatino
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jesse J Salk
- Department of Medicine, Divisions of Hematology and Medical Oncology, University of Washington School of Medicine, Seattle, Washington, USA
- TwinStrand Biosciences Inc., Seattle, Washington, USA
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael Themis
- TestaVec Ltd, Maidenhead, United Kingdom
- Division of Biosciences, Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Jing Yuan
- Kymera Therapeutics, Watertown, Massachusetts, USA
| |
Collapse
|
5
|
Anderson JS, Lodigiani AL, Barbaduomo CM, Beegle JR. Hematopoietic stem cell gene therapy for the treatment of SYNGAP1-related non-specific intellectual disability. J Gene Med 2024; 26:e3717. [PMID: 38967915 DOI: 10.1002/jgm.3717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/15/2024] [Accepted: 06/02/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND Synaptic Ras GTPase activating protein 1 (SYNGAP1)-related non-specific intellectual disability is a neurodevelopmental disorder caused by an insufficient level of SynGAP1 resulting in a dysfunction of neuronal synapses and presenting with a wide array of clinical phenotypes. Hematopoietic stem cell gene therapy has the potential to deliver therapeutic levels of functional SynGAP1 to affected neurons upon transduction of hematopoietic stem and progenitor cells with a lentiviral vector. METHODS As a novel approach toward the treatment of SYNGAP1, we have generated a lentiviral vector expressing a modified form of SynGAP1 for transduction of human CD34+ hematopoietic stem and progenitor cells. The gene-modified cells were then transplanted into adult immunodeficient SYNGAP1+/- heterozygous mice and evaluated for improvement of SYNGAP1-related clinical phenotypes. Expression of SynGAP1 was also evaluated in the brain tissue of transplanted mice. RESULTS In our proof-of-concept study, we have demonstrated significant improvement of SYNGAP1-related phenotypes including an improvement in motor abilities observed in mice transplanted with the vector transduced cells because they displayed decreased hyperactivity in an open field assay and an increased latency to fall in a rotarod assay. An increased level of SynGAP1 was also detected in the brains of these mice. CONCLUSIONS These early-stage results highlight the potential of this stem cell gene therapy approach as a treatment strategy for SYNGAP1.
Collapse
Affiliation(s)
- Joseph S Anderson
- Department of Internal Medicine, University of California Davis, Sacramento, CA, USA
| | - Alyse L Lodigiani
- Department of Internal Medicine, University of California Davis, Sacramento, CA, USA
| | - Camilla M Barbaduomo
- Department of Internal Medicine, University of California Davis, Sacramento, CA, USA
| | - Julie R Beegle
- Department of Internal Medicine, University of California Davis, Sacramento, CA, USA
| |
Collapse
|
6
|
Ago Y, Rintz E, Musini KS, Ma Z, Tomatsu S. Molecular Mechanisms in Pathophysiology of Mucopolysaccharidosis and Prospects for Innovative Therapy. Int J Mol Sci 2024; 25:1113. [PMID: 38256186 PMCID: PMC10816168 DOI: 10.3390/ijms25021113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Mucopolysaccharidoses (MPSs) are a group of inborn errors of the metabolism caused by a deficiency in the lysosomal enzymes required to break down molecules called glycosaminoglycans (GAGs). These GAGs accumulate over time in various tissues and disrupt multiple biological systems, including catabolism of other substances, autophagy, and mitochondrial function. These pathological changes ultimately increase oxidative stress and activate innate immunity and inflammation. We have described the pathophysiology of MPS and activated inflammation in this paper, starting with accumulating the primary storage materials, GAGs. At the initial stage of GAG accumulation, affected tissues/cells are reversibly affected but progress irreversibly to: (1) disruption of substrate degradation with pathogenic changes in lysosomal function, (2) cellular dysfunction, secondary/tertiary accumulation (toxins such as GM2 or GM3 ganglioside, etc.), and inflammatory process, and (3) progressive tissue/organ damage and cell death (e.g., skeletal dysplasia, CNS impairment, etc.). For current and future treatment, several potential treatments for MPS that can penetrate the blood-brain barrier and bone have been proposed and/or are in clinical trials, including targeting peptides and molecular Trojan horses such as monoclonal antibodies attached to enzymes via receptor-mediated transport. Gene therapy trials with AAV, ex vivo LV, and Sleeping Beauty transposon system for MPS are proposed and/or underway as innovative therapeutic options. In addition, possible immunomodulatory reagents that can suppress MPS symptoms have been summarized in this review.
Collapse
Affiliation(s)
- Yasuhiko Ago
- Nemours Children’s Health, 1600 Rockland Rd., Wilmington, DE 19803, USA; (Y.A.); (K.S.M.); (Z.M.)
| | - Estera Rintz
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, 80-308 Gdansk, Poland;
| | - Krishna Sai Musini
- Nemours Children’s Health, 1600 Rockland Rd., Wilmington, DE 19803, USA; (Y.A.); (K.S.M.); (Z.M.)
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Zhengyu Ma
- Nemours Children’s Health, 1600 Rockland Rd., Wilmington, DE 19803, USA; (Y.A.); (K.S.M.); (Z.M.)
| | - Shunji Tomatsu
- Nemours Children’s Health, 1600 Rockland Rd., Wilmington, DE 19803, USA; (Y.A.); (K.S.M.); (Z.M.)
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu 501-1112, Japan
- Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA 19144, USA
| |
Collapse
|
7
|
Liu S. Lentiviral Production Platform. Methods Mol Biol 2024; 2766:163-168. [PMID: 38270876 DOI: 10.1007/978-1-0716-3682-4_17] [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] [Indexed: 01/26/2024]
Abstract
Lentiviral-mediated transfection technique is a powerful tool for gene modification in preclinical studies. By using this technique, the desired gene modification can be achieved easily in immune cells, nondividing, and terminally differentiated cells, including hematopoietic stem cells, neurons, and even tumor cells, which other viral vectors cannot do. The main considerations of therapeutic gene delivery using a lentiviral system are the risk of insertional mutagenesis and the immune reaction elicited by infected cells. Although some biosafety concerns need to be addressed before clinical trials in rheumatoid arthritis, the lentiviral system targeting therapeutic targets has been widely used for in vivo gene transfer in animal models. In this chapter, the protocols for production of viral particles and viral concentration are provided. As an alternative utilization, this lentiviral production platform could also be employed to produce a pseudotype severe acute respiratory syndrome-related coronavirus 2 in which the spike glycoprotein of SARS-CoV-2 was incorporated into pseudovirions for viral study.
Collapse
Affiliation(s)
- Shuang Liu
- Department of Pharmacology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan.
| |
Collapse
|
8
|
Rose SC, Larsen M, Xie Y, Sharfstein ST. Salivary Gland Bioengineering. Bioengineering (Basel) 2023; 11:28. [PMID: 38247905 PMCID: PMC10813147 DOI: 10.3390/bioengineering11010028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/19/2023] [Accepted: 11/30/2023] [Indexed: 01/23/2024] Open
Abstract
Salivary gland dysfunction affects millions globally, and tissue engineering may provide a promising therapeutic avenue. This review delves into the current state of salivary gland tissue engineering research, starting with a study of normal salivary gland development and function. It discusses the impact of fibrosis and cellular senescence on salivary gland pathologies. A diverse range of cells suitable for tissue engineering including cell lines, primary salivary gland cells, and stem cells are examined. Moreover, the paper explores various supportive biomaterials and scaffold fabrication methodologies that enhance salivary gland cell survival, differentiation, and engraftment. Innovative engineering strategies for the improvement of vascularization, innervation, and engraftment of engineered salivary gland tissue, including bioprinting, microfluidic hydrogels, mesh electronics, and nanoparticles, are also evaluated. This review underscores the promising potential of this research field for the treatment of salivary gland dysfunction and suggests directions for future exploration.
Collapse
Affiliation(s)
- Stephen C. Rose
- Department of Nanoscale Science and Engineering, College of Nanotechnology, Science, and Engineering, University at Albany, SUNY, 257 Fuller Road, Albany, NY 12203, USA (Y.X.)
| | - Melinda Larsen
- Department of Biological Sciences and The RNA Institute, University at Albany, SUNY, 1400 Washington Ave., Albany, NY 12222, USA;
| | - Yubing Xie
- Department of Nanoscale Science and Engineering, College of Nanotechnology, Science, and Engineering, University at Albany, SUNY, 257 Fuller Road, Albany, NY 12203, USA (Y.X.)
| | - Susan T. Sharfstein
- Department of Nanoscale Science and Engineering, College of Nanotechnology, Science, and Engineering, University at Albany, SUNY, 257 Fuller Road, Albany, NY 12203, USA (Y.X.)
| |
Collapse
|
9
|
Breda L, Papp TE, Triebwasser MP, Yadegari A, Fedorky MT, Tanaka N, Abdulmalik O, Pavani G, Wang Y, Grupp SA, Chou ST, Ni H, Mui BL, Tam YK, Weissman D, Rivella S, Parhiz H. In vivo hematopoietic stem cell modification by mRNA delivery. Science 2023; 381:436-443. [PMID: 37499029 PMCID: PMC10567133 DOI: 10.1126/science.ade6967] [Citation(s) in RCA: 100] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 06/01/2023] [Indexed: 07/29/2023]
Abstract
Hematopoietic stem cells (HSCs) are the source of all blood cells over an individual's lifetime. Diseased HSCs can be replaced with gene-engineered or healthy HSCs through HSC transplantation (HSCT). However, current protocols carry major side effects and have limited access. We developed CD117/LNP-messenger RNA (mRNA), a lipid nanoparticle (LNP) that encapsulates mRNA and is targeted to the stem cell factor receptor (CD117) on HSCs. Delivery of the anti-human CD117/LNP-based editing system yielded near-complete correction of hematopoietic sickle cells. Furthermore, in vivo delivery of pro-apoptotic PUMA (p53 up-regulated modulator of apoptosis) mRNA with CD117/LNP affected HSC function and permitted nongenotoxic conditioning for HSCT. The ability to target HSCs in vivo offers a nongenotoxic conditioning regimen for HSCT, and this platform could be the basis of in vivo genome editing to cure genetic disorders, which would abrogate the need for HSCT.
Collapse
Affiliation(s)
- Laura Breda
- Department of Pediatrics, Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Tyler E Papp
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael P Triebwasser
- Department of Pediatrics, Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, The University of Michigan, Ann Arbor, MI, USA
| | - Amir Yadegari
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Megan T Fedorky
- Department of Pediatrics, Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Naoto Tanaka
- Department of Pediatrics, Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Osheiza Abdulmalik
- Department of Pediatrics, Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Giulia Pavani
- Department of Pathology and Laboratory Medicine, Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Yongping Wang
- Department of Pathology and Laboratory Medicine, Transfusion Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Clinical Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Stephan A Grupp
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Departments of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stella T Chou
- Department of Pediatrics, Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Transfusion Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Houping Ni
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Ying K Tam
- Acuitas Therapeutics, Vancouver, BC V6T1Z3, Canada
| | - Drew Weissman
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stefano Rivella
- Department of Pediatrics, Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Cell and Molecular Biology affinity group, University of Pennsylvania, Philadelphia, PA, USA
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Penn Center for Musculoskeletal Disorders, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Penn Institute for RNA Innovation, University of Pennsylvania, Philadelphia, PA, USA
| | - Hamideh Parhiz
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
10
|
Iancu O, Allen D, Knop O, Zehavi Y, Breier D, Arbiv A, Lev A, Lee YN, Beider K, Nagler A, Somech R, Hendel A. Multiplex HDR for disease and correction modeling of SCID by CRISPR genome editing in human HSPCs. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 31:105-121. [PMID: 36618262 PMCID: PMC9813580 DOI: 10.1016/j.omtn.2022.12.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022]
Abstract
Severe combined immunodeficiency (SCID) is a group of disorders caused by mutations in genes involved in the process of lymphocyte maturation and function. CRISPR-Cas9 gene editing of the patient's own hematopoietic stem and progenitor cells (HSPCs) ex vivo could provide a therapeutic alternative to allogeneic hematopoietic stem cell transplantation, the current gold standard for treatment of SCID. To eliminate the need for scarce patient samples, we engineered genotypes in healthy donor (HD)-derived CD34+ HSPCs using CRISPR-Cas9/rAAV6 gene-editing, to model both SCID and the therapeutic outcomes of gene-editing therapies for SCID via multiplexed homology-directed repair (HDR). First, we developed a SCID disease model via biallelic knockout of genes critical to the development of lymphocytes; and second, we established a knockin/knockout strategy to develop a proof-of-concept single-allelic gene correction. Based on these results, we performed gene correction of RAG2-SCID patient-derived CD34+ HSPCs that successfully developed into CD3+ T cells with diverse TCR repertoires in an in vitro T cell differentiation platform. In summary, we present a strategy to determine the optimal configuration for CRISPR-Cas9 gene correction of SCID using HD-derived CD34+ HSPCs, and the feasibility of translating this gene correction approach in patient-derived CD34+ HSPCs.
Collapse
Affiliation(s)
- Ortal Iancu
- The Institute for Advanced Materials and Nanotechnology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Daniel Allen
- The Institute for Advanced Materials and Nanotechnology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Orli Knop
- The Institute for Advanced Materials and Nanotechnology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Yonathan Zehavi
- The Institute for Advanced Materials and Nanotechnology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Dor Breier
- The Institute for Advanced Materials and Nanotechnology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Adaya Arbiv
- The Institute for Advanced Materials and Nanotechnology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Atar Lev
- The Institute for Advanced Materials and Nanotechnology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-HaShomer, Ramat Gan 5266202, Israel
| | - Yu Nee Lee
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-HaShomer, Ramat Gan 5266202, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Katia Beider
- The Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-HaShomer, Ramat Gan 5266202, Israel
| | - Arnon Nagler
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- The Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-HaShomer, Ramat Gan 5266202, Israel
| | - Raz Somech
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-HaShomer, Ramat Gan 5266202, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ayal Hendel
- The Institute for Advanced Materials and Nanotechnology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| |
Collapse
|
11
|
Platani M, Sokefun O, Bassil E, Apidianakis Y. Genetic engineering and genome editing in plants, animals and humans: Facts and myths. Gene 2023; 856:147141. [PMID: 36574935 DOI: 10.1016/j.gene.2022.147141] [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: 03/30/2022] [Revised: 12/17/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022]
Abstract
Human history is inextricably linked to the introduction of desirable heritable traits in plants and animals. Selective breeding (SB) predates our historical period and has been practiced since the advent of agriculture and farming more than ten thousand years ago. Since the 1970s, methods of direct plant and animal genome manipulation are constantly being developed. These are collectively described as "genetic engineering" (GE). Plant GE aims to improve nutritional value, insect resistance and weed control. Animal GE has focused on livestock improvement and disease control. GE applications also involve medical improvements intended to treat human disease. The scientific consensus built around marketed products of GE organisms (GEOs) is usually well established, noting significant benefits and low risks. GEOs are exhaustively scrutinized in the EU and many non-EU countries for their effects on human health and the environment, but scrutiny should be equally applied to all previously untested organisms derived directly from nature or through selective breeding. In fact, there is no evidence to suggest that natural or selectively bred plants and animals are in principle safer to humans than GEOs. Natural and selectively bred strains evolve over time via genetic mutations that can be as risky to humans and the environment as the mutations found in GEOs. Thus, previously untested plant and animal strains aimed for marketing should be proven useful or harmful to humans only upon comparative testing, regardless of their origin. Highlighting the scientific consensus declaring significant benefits and rather manageable risks provided by equitably accessed GEOs, can mitigate negative predispositions by policy makers and the public. Accordingly, we provide an overview of the underlying technologies and the scientific consensus to help resolve popular myths about the safety and usefulness of GEOs.
Collapse
Affiliation(s)
- Maria Platani
- Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
| | - Owolabi Sokefun
- Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
| | - Elias Bassil
- Horticultural Sciences Department, University of Florida, Gainesville, USA
| | | |
Collapse
|
12
|
Castiello MC, Ferrari S, Villa A. Correcting inborn errors of immunity: From viral mediated gene addition to gene editing. Semin Immunol 2023; 66:101731. [PMID: 36863140 PMCID: PMC10109147 DOI: 10.1016/j.smim.2023.101731] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 01/25/2023] [Accepted: 02/14/2023] [Indexed: 03/04/2023]
Abstract
Allogeneic hematopoietic stem cell transplantation is an effective treatment to cure inborn errors of immunity. Remarkable progress has been achieved thanks to the development and optimization of effective combination of advanced conditioning regimens and use of immunoablative/suppressive agents preventing rejection as well as graft versus host disease. Despite these tremendous advances, autologous hematopoietic stem/progenitor cell therapy based on ex vivo gene addition exploiting integrating γ-retro- or lenti-viral vectors, has demonstrated to be an innovative and safe therapeutic strategy providing proof of correction without the complications of the allogeneic approach. The recent advent of targeted gene editing able to precisely correct genomic variants in an intended locus of the genome, by introducing deletions, insertions, nucleotide substitutions or introducing a corrective cassette, is emerging in the clinical setting, further extending the therapeutic armamentarium and offering a cure to inherited immune defects not approachable by conventional gene addition. In this review, we will analyze the current state-of-the art of conventional gene therapy and innovative protocols of genome editing in various primary immunodeficiencies, describing preclinical models and clinical data obtained from different trials, highlighting potential advantages and limits of gene correction.
Collapse
Affiliation(s)
- Maria Carmina Castiello
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy; Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (IRGB-CNR), Milan, Italy
| | - Samuele Ferrari
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy; Vita-Salute San Raffaele University, Milan 20132, Italy
| | - Anna Villa
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy; Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (IRGB-CNR), Milan, Italy.
| |
Collapse
|
13
|
van Heuvel Y, Schatz S, Hein M, Dogra T, Kazenmaier D, Tschorn N, Genzel Y, Stitz J. Novel suspension retroviral packaging cells generated by transposition using transposase encoding mRNA advance vector yields and enable production in bioreactors. Front Bioeng Biotechnol 2023; 11:1076524. [PMID: 37082212 PMCID: PMC10112512 DOI: 10.3389/fbioe.2023.1076524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 03/24/2023] [Indexed: 04/22/2023] Open
Abstract
To date, the establishment of high-titer stable viral packaging cells (VPCs) at large scale for gene therapeutic applications is very time- and cost-intensive. Here we report the establishment of three human suspension 293-F-derived ecotropic MLV-based VPCs. The classic stable transfection of an EGFP-expressing transfer vector resulted in a polyclonal VPC pool that facilitated cultivation in shake flasks of 100 mL volumes and yielded high functional titers of more than 1 × 106 transducing units/mL (TU/mL). When the transfer vector was flanked by transposon terminal inverted repeats (TIRs) and upon co-transfection of a plasmid encoding for the transposase, productivities could be slightly elevated to more than 3 × 106 TU/mL. In contrast and using mRNA encoding for the transposase, as a proof of concept, productivities were drastically improved by more than ten-fold exceeding 5 × 107 TU/mL. In addition, these VPC pools were generated within only 3 weeks. The production volume was successfully scaled up to 500 mL employing a stirred-tank bioreactor (STR). We anticipate that the stable transposition of transfer vectors employing transposase transcripts will be of utility for the future establishment of high-yield VPCs producing pseudotype vector particles with a broader host tropism on a large scale.
Collapse
Affiliation(s)
- Yasemin van Heuvel
- Research Group Medical Biotechnology and Bioengineering, Faculty of Applied Natural Sciences, University of Applied Sciences Cologne, Campus Leverkusen, Cologne, Germany
- Institute of Technical Chemistry, Gottfried Wilhelm Leibniz University Hannover, Hanover, Germany
| | - Stefanie Schatz
- Research Group Medical Biotechnology and Bioengineering, Faculty of Applied Natural Sciences, University of Applied Sciences Cologne, Campus Leverkusen, Cologne, Germany
- Institute of Technical Chemistry, Gottfried Wilhelm Leibniz University Hannover, Hanover, Germany
| | - Marc Hein
- Chair of Bioprocess Engineering, Otto-Von-Guericke-University Magdeburg, Magdeburg, Germany
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Tanya Dogra
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Daniel Kazenmaier
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
- Faculty of Biotechnology, University of Applied Sciences Mannheim, Mannheim, Germany
| | - Natalie Tschorn
- Research Group Medical Biotechnology and Bioengineering, Faculty of Applied Natural Sciences, University of Applied Sciences Cologne, Campus Leverkusen, Cologne, Germany
- Institute of Technical Chemistry, Gottfried Wilhelm Leibniz University Hannover, Hanover, Germany
| | - Yvonne Genzel
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Jörn Stitz
- Research Group Medical Biotechnology and Bioengineering, Faculty of Applied Natural Sciences, University of Applied Sciences Cologne, Campus Leverkusen, Cologne, Germany
- *Correspondence: Jörn Stitz,
| |
Collapse
|
14
|
Kim Y, Kim H, Kim EH, Jang H, Jang Y, Chi SG, Yang Y, Kim SH. The Potential of Cell-Penetrating Peptides for mRNA Delivery to Cancer Cells. Pharmaceutics 2022; 14:pharmaceutics14061271. [PMID: 35745843 PMCID: PMC9227323 DOI: 10.3390/pharmaceutics14061271] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 02/01/2023] Open
Abstract
In vitro transcribed mRNA for the synthesis of any given protein has shown great potential in cancer gene therapy, especially in cancer vaccines for immunotherapy. To overcome physiological barriers, such as rapid degradation by enzymatic attack and poor cellular uptake due to their large size and hydrophilic properties, many delivery carriers for mRNAs are being investigated for improving the bioavailability of mRNA. Recently, cell-penetrating peptides (CPPs) have received attention as promising tools for gene delivery. In terms of their biocompatibility and the ability to target specific cells with the versatility of peptide sequences, they may provide clues to address the challenges of conventional delivery systems for cancer mRNA delivery. In this study, optimal conditions for the CPP/mRNA complexes were identified in terms of complexation capacity and N/P ratio, and protection against RNase was confirmed. When cancer cells were treated at a concentration of 6.8 nM, which could deliver the highest amount of mRNA without toxicity, the amphipathic CPP/mRNA complexes with a size less than 200 nm showed high cellular uptake and protein expression. With advances in our understanding of CPPs, CPPs designed to target tumor tissues will be promising for use in developing a new class of mRNA delivery vehicles in cancer therapy.
Collapse
Affiliation(s)
- Yelee Kim
- Medical Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (Y.K.); (H.K.); (E.H.K.); (H.J.); (Y.J.)
- Department of Life Sciences, Korea University, Seoul 02841, Korea;
| | - Hyosuk Kim
- Medical Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (Y.K.); (H.K.); (E.H.K.); (H.J.); (Y.J.)
| | - Eun Hye Kim
- Medical Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (Y.K.); (H.K.); (E.H.K.); (H.J.); (Y.J.)
- Department of Life Sciences, Korea University, Seoul 02841, Korea;
| | - Hochung Jang
- Medical Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (Y.K.); (H.K.); (E.H.K.); (H.J.); (Y.J.)
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Korea
| | - Yeongji Jang
- Medical Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (Y.K.); (H.K.); (E.H.K.); (H.J.); (Y.J.)
- Department of Life Sciences, Korea University, Seoul 02841, Korea;
| | - Sung-Gil Chi
- Department of Life Sciences, Korea University, Seoul 02841, Korea;
| | - Yoosoo Yang
- Medical Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (Y.K.); (H.K.); (E.H.K.); (H.J.); (Y.J.)
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Korea
- Correspondence: (Y.Y.); (S.H.K.); Tel.: +82-2-958-6655 (Y.Y.); +82-2-958-6639 (S.H.K.)
| | - Sun Hwa Kim
- Medical Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (Y.K.); (H.K.); (E.H.K.); (H.J.); (Y.J.)
- Correspondence: (Y.Y.); (S.H.K.); Tel.: +82-2-958-6655 (Y.Y.); +82-2-958-6639 (S.H.K.)
| |
Collapse
|
15
|
Cheng A, Harikrishna JA, Redwood CS, Lit LC, Nath SK, Chua KH. Genetics Matters: Voyaging from the Past into the Future of Humanity and Sustainability. Int J Mol Sci 2022; 23:ijms23073976. [PMID: 35409335 PMCID: PMC8999725 DOI: 10.3390/ijms23073976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/21/2022] [Accepted: 03/30/2022] [Indexed: 12/02/2022] Open
Abstract
The understanding of how genetic information may be inherited through generations was established by Gregor Mendel in the 1860s when he developed the fundamental principles of inheritance. The science of genetics, however, began to flourish only during the mid-1940s when DNA was identified as the carrier of genetic information. The world has since then witnessed rapid development of genetic technologies, with the latest being genome-editing tools, which have revolutionized fields from medicine to agriculture. This review walks through the historical timeline of genetics research and deliberates how this discipline might furnish a sustainable future for humanity.
Collapse
Affiliation(s)
- Acga Cheng
- Institute of Biological Science, Faculty of Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (A.C.); (J.A.H.)
| | - Jennifer Ann Harikrishna
- Institute of Biological Science, Faculty of Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (A.C.); (J.A.H.)
- Centre for Research in Biotechnology for Agriculture, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Charles S. Redwood
- Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK;
| | - Lei Cheng Lit
- Department of Physiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
| | - Swapan K. Nath
- Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Correspondence: (S.K.N.); (K.H.C.)
| | - Kek Heng Chua
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
- Correspondence: (S.K.N.); (K.H.C.)
| |
Collapse
|
16
|
Pizevska M, Kaeda J, Fritsche E, Elazaly H, Reinke P, Amini L. Advanced Therapy Medicinal Products' Translation in Europe: A Developers' Perspective. Front Med (Lausanne) 2022; 9:757647. [PMID: 35186986 PMCID: PMC8851388 DOI: 10.3389/fmed.2022.757647] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 01/10/2022] [Indexed: 11/13/2022] Open
Abstract
Advanced Therapy Medicinal Products (ATMPs) comprising cell, gene, and tissue-engineered therapies have demonstrated enormous therapeutic benefits. However, their development is complex to be managed efficiently within currently existing regulatory frameworks. Legislation and regulation requirements for ATMPs must strike a balance between the patient safety while promoting innovations to optimize exploitation of these novel therapeutics. This paradox highlights the importance of on-going dynamic dialogue between all stakeholders and regulatory science to facilitate the development of pragmatic ATMP regulatory guidelines.
Collapse
Affiliation(s)
- Maja Pizevska
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin Institute of Health Center for Regenerative Therapies, Berlin, Germany
| | - Jaspal Kaeda
- Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Enrico Fritsche
- Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Hisham Elazaly
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin Institute of Health Center for Regenerative Therapies, Berlin, Germany
| | - Petra Reinke
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin Institute of Health Center for Regenerative Therapies, Berlin, Germany.,Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Leila Amini
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin Institute of Health Center for Regenerative Therapies, Berlin, Germany.,Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
17
|
Braga LAM, Conte Filho CG, Mota FB. Future of genetic therapies for rare genetic diseases: what to expect for the next 15 years? THERAPEUTIC ADVANCES IN RARE DISEASE 2022; 3:26330040221100840. [PMID: 37180410 PMCID: PMC10032453 DOI: 10.1177/26330040221100840] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 04/22/2022] [Indexed: 05/16/2023]
Abstract
Introduction Rare genetic diseases affect millions of people worldwide. Most of them are caused by defective genes that impair quality of life and can lead to premature death. As genetic therapies aim to fix or replace defective genes, they are considered the most promising treatment for rare genetic diseases. Yet, as these therapies are still under development, it is still unclear whether they will be successful in treating these diseases. This study aims to address this gap by assessing researchers' opinions on the future of genetic therapies for the treatment of rare genetic diseases. Methods We conducted a global cross-sectional web-based survey of researchers who recently authored peer-reviewed articles related to rare genetic diseases. Results We assessed the opinions of 1430 researchers with high and good knowledge about genetic therapies for the treatment of rare genetic diseases. Overall, the respondents believed that genetic therapies would be the standard of care for rare genetic diseases before 2036, leading to cures after this period. CRISPR-Cas9 was considered the most likely approach to fixing or replacing defective genes in the next 15 years. The respondents with good knowledge believed that genetic therapies would only have long-lasting effects after 2036, while those with high knowledge were divided on this issue. The respondents with good knowledge on the subject believed that non-viral vectors are more likely to be successful in fixing or replacing defective genes in the next 15 years, while most of the respondents with high knowledge believed viral vectors would be more successful. Conclusion Overall, the researchers who participated in this study expect that in the future genetic therapies will greatly benefit the treatment of patients with rare genetic diseases.
Collapse
Affiliation(s)
| | | | - Fabio Batista Mota
- Laboratory of Cellular Communication, Oswaldo
Cruz Institute, Oswaldo Cruz Foundation, Av. Brasil, 4.365, Pavilhão 108,
Manguinhos, Rio de Janeiro RJ 21040-360, Brazil
| |
Collapse
|
18
|
Development and clinical translation of ex vivo gene therapy. Comput Struct Biotechnol J 2022; 20:2986-3003. [PMID: 35782737 PMCID: PMC9218169 DOI: 10.1016/j.csbj.2022.06.015] [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: 02/08/2022] [Revised: 06/07/2022] [Accepted: 06/07/2022] [Indexed: 11/27/2022] Open
Abstract
Retroviral gene therapy has emerged as a promising therapeutic modality for multiple inherited and acquired human diseases. The capability of delivering curative treatment or mediating therapeutic benefits for a long-term period following a single application fundamentally distinguishes this medical intervention from traditional medicine and various lentiviral/γ-retroviral vector-mediated gene therapy products have been approved for clinical use. Continued advances in retroviral vector engineering, genomic editing, synthetic biology and immunology will broaden the medical applications of gene therapy and improve the efficacy and safety of the treatments based on genetic correction and alteration. This review will summarize the advent and clinical translation of ex vivo gene therapy, with the focus on the milestones during the exploitation of genetically engineered hematopoietic stem cells (HSCs) tackling a variety of pathological conditions which led to marketing approval. Finally, current statue and future prospects of gene editing as an alternative therapeutic approach are also discussed.
Collapse
|
19
|
Ghosh P, Bhattacharya M, Patra P, Sharma G, Patra BC, Lee SS, Sharma AR, Chakraborty C. Evaluation and Designing of Epitopic-Peptide Vaccine Against Bunyamwera orthobunyavirus Using M-Polyprotein Target Sequences. Int J Pept Res Ther 2021; 28:5. [PMID: 34867129 PMCID: PMC8634745 DOI: 10.1007/s10989-021-10322-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2021] [Indexed: 11/30/2022]
Abstract
Bunyamwera orthobunyavirus and its serogroup can cause several diseases in humans, cattle, ruminants, and birds. The viral M-polyprotein helps the virus to enter the host body. Therefore, this protein might serve as a potential vaccine target against Bunyamwera orthobunyavirus. The present study applied the immunoinformatics technique to design an epitopic vaccine component that could protect against Bunyamwera infection. Phylogenetic analysis revealed the presence of conserved patterns of M-polyprotein within the viral serogroup. Three epitopes common for both B-cell and T-cell were identified, i.e., YQPTELTRS, YKAHDKEET, and ILGTGTPKF merged with a specific linker peptide to construct an active vaccine component. The low atomic contact energy value of docking complex between human TLR4 (TLR4/MD2 complex) and vaccine construct confirms the elevated protein–protein binding interaction. Molecular dynamic simulation and normal mode analysis illustrate the docking complex’s stability, especially by the higher Eigenvalue. In silico cloning of the vaccine construct was applied to amplify the desired vaccine component. Structural allocation of both the vaccine and epitopes also show the efficacy of the developed vaccine. Hence, the computational research design outcomes support that the peptide-based vaccine construction is a crucial drive target to limit the infection of Bunyamwera orthobunyavirus to an extent.
Collapse
Affiliation(s)
- Pratik Ghosh
- Department of Zoology, Vidyasagar University, Midnapore, West Bengal 721102 India
| | - Manojit Bhattacharya
- Department of Zoology, Fakir Mohan University, Vyasa Vihar, Balasore, Odisha 756020 India
| | - Prasanta Patra
- Department of Zoology, Vidyasagar University, Midnapore, West Bengal 721102 India
| | - Garima Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chuncheon-si, Republic of Korea
| | - Bidhan Chandra Patra
- Department of Zoology, Vidyasagar University, Midnapore, West Bengal 721102 India
| | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, 24252 Gangwon-do Republic of Korea
| | - Ashish Ranjan Sharma
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, 24252 Gangwon-do Republic of Korea
| | - Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Barasat-Barrackpore Rd, Kolkata, West Bengal 700126 India
| |
Collapse
|
20
|
Vicente MM, Chaves-Ferreira M, Jorge JMP, Proença JT, Barreto VM. The Off-Targets of Clustered Regularly Interspaced Short Palindromic Repeats Gene Editing. Front Cell Dev Biol 2021; 9:718466. [PMID: 34604217 PMCID: PMC8484971 DOI: 10.3389/fcell.2021.718466] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/09/2021] [Indexed: 12/26/2022] Open
Abstract
The repurposing of the CRISPR/Cas bacterial defense system against bacteriophages as simple and flexible molecular tools has revolutionized the field of gene editing. These tools are now widely used in basic research and clinical trials involving human somatic cells. However, a global moratorium on all clinical uses of human germline editing has been proposed because the technology still lacks the required efficacy and safety. Here we focus on the approaches developed since 2013 to decrease the frequency of unwanted mutations (the off-targets) during CRISPR-based gene editing.
Collapse
Affiliation(s)
- Manuel M Vicente
- DNA Breaks Group, NOVA Medical School (NMS), Centro de Estudos de Doenças Crónicas (CEDOC), NOVA University of Lisbon, Lisbon, Portugal
| | - Miguel Chaves-Ferreira
- DNA Breaks Group, NOVA Medical School (NMS), Centro de Estudos de Doenças Crónicas (CEDOC), NOVA University of Lisbon, Lisbon, Portugal
| | - João M P Jorge
- DNA Breaks Group, NOVA Medical School (NMS), Centro de Estudos de Doenças Crónicas (CEDOC), NOVA University of Lisbon, Lisbon, Portugal
| | - João T Proença
- DNA Breaks Group, NOVA Medical School (NMS), Centro de Estudos de Doenças Crónicas (CEDOC), NOVA University of Lisbon, Lisbon, Portugal
| | - Vasco M Barreto
- DNA Breaks Group, NOVA Medical School (NMS), Centro de Estudos de Doenças Crónicas (CEDOC), NOVA University of Lisbon, Lisbon, Portugal
| |
Collapse
|
21
|
Fu X, Chen T, Song Y, Feng C, Chen H, Zhang Q, Chen G, Zhu X. mRNA Delivery by a pH-Responsive DNA Nano-Hydrogel. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2101224. [PMID: 34145748 DOI: 10.1002/smll.202101224] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Indexed: 06/12/2023]
Abstract
The delivery of mRNA to manipulate protein expression has attracted widespread attention, since that mRNA overcomes the problem of infection and mutation risks in transgenes and can work as drugs for the treatment of diseases. Although there are currently some vehicles that deliver mRNA into cells, they have not yet reached a good balance in terms of expression efficiency and biocompatibility. Here, a DNA nano-hydrogel system for mRNA delivery is developed. The nano-hydrogel is all composed of DNA except the target mRNA, so it has superior biocompatibility compared with those chemical vehicles. In parallel, the nano-hydrogel can be compacted into a nanosphere under the crosslinking by well-designed "X"-shaped DNA scaffolds and DNA linkers, facilitating the delivery into cells through endocytosis. In addition, smart intracellular release of the mRNA is achieved by incorporating a pH-responsive i-motif structure into the nano-hydrogel. Thus, taking the efficient delivery and release together, mRNA can be translated into the corresponding protein with a high efficiency, which is comparable to that of the commercial liposome but with a much better biocompatibility. Due to the excellent biocompatibility and efficiency, this nano-hydrogel system is expected to become a competitive alternative for delivering functional mRNA in vivo.
Collapse
Affiliation(s)
- Xin Fu
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Tianshu Chen
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Yuchen Song
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Chang Feng
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Huinan Chen
- College of Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Qianqian Zhang
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Guifang Chen
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Xiaoli Zhu
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| |
Collapse
|
22
|
Kansal AR, Reifsnider OS, Brand SB, Hawkins N, Coughlan A, Li S, Cragin L, Paramore C, Dietz AC, Caro JJ. Economic evaluation of betibeglogene autotemcel (Beti-cel) gene addition therapy in transfusion-dependent β-thalassemia. JOURNAL OF MARKET ACCESS & HEALTH POLICY 2021; 9:1922028. [PMID: 34178295 PMCID: PMC8205006 DOI: 10.1080/20016689.2021.1922028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/19/2021] [Accepted: 04/21/2021] [Indexed: 05/22/2023]
Abstract
Background: Standard of care (SoC) for transfusion-dependent β-thalassemia (TDT) requires lifelong, regular blood transfusions as well as chelation to reduce iron accumulation. Objective: This study investigates the cost-effectiveness of betibeglogene autotemcel ('beti-cel'; LentiGlobin for β-thalassemia) one-time, gene addition therapy compared to lifelong SoC for TDT. Study design: Microsimulation model simulated the lifetime course of TDT based on a causal sequence in which transfusion requirements determine tissue iron levels, which in turn determine risk of iron overload complications that increase mortality. Clinical trial data informed beti-cel clinical parameters; effects of SoC on iron levels came from real-world studies; iron overload complication rates and mortality were based on published literature. Setting: USA; commercial payer perspective Participants: TDT patients age 2-50 Interventions: Beti-cel is compared to SoC. Main outcome measure: Incremental cost-effectiveness ratio (ICER) utilizing quality-adjusted life-years (QALYs) Results: The model predicts beti-cel adds 3.8 discounted life years (LYs) or 6.9 QALYs versus SoC. Discounted lifetime costs were $2.28 M for beti-cel ($572,107 if excluding beti-cel cost) and $2.04 M for SoC, with a resulting ICER of $34,833 per QALY gained. Conclusion: Beti-cel is cost-effective for TDT patients compared to SoC. This is due to longer survival and cost offset of lifelong SoC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - J. Jaime Caro
- Evidera, Inc., Waltham, MA, USA
- CONTACT J. Jaime Caro Evidera, Inc., Waltham, MA, USA
| |
Collapse
|
23
|
Yoder KE, Rabe AJ, Fishel R, Larue RC. Strategies for Targeting Retroviral Integration for Safer Gene Therapy: Advances and Challenges. Front Mol Biosci 2021; 8:662331. [PMID: 34055882 PMCID: PMC8149907 DOI: 10.3389/fmolb.2021.662331] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/29/2021] [Indexed: 12/11/2022] Open
Abstract
Retroviruses are obligate intracellular parasites that must integrate a copy of the viral genome into the host DNA. The integration reaction is performed by the viral enzyme integrase in complex with the two ends of the viral cDNA genome and yields an integrated provirus. Retroviral vector particles are attractive gene therapy delivery tools due to their stable integration. However, some retroviral integration events may dysregulate host oncogenes leading to cancer in gene therapy patients. Multiple strategies to target retroviral integration, particularly to genetic safe harbors, have been tested with limited success. Attempts to target integration may be limited by the multimerization of integrase or the presence of host co-factors for integration. Several retroviral integration complexes have evolved a mechanism of tethering to chromatin via a host protein. Integration host co-factors bind chromatin, anchoring the complex and allowing integration. The tethering factor allows for both close proximity to the target DNA and specificity of targeting. Each retrovirus appears to have distinct preferences for DNA sequence and chromatin features at the integration site. Tethering factors determine the preference for chromatin features, but do not affect the subtle sequence preference at the integration site. The sequence preference is likely intrinsic to the integrase protein. New developments may uncouple the requirement for a tethering factor and increase the ability to redirect retroviral integration.
Collapse
Affiliation(s)
- Kristine E Yoder
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Anthony J Rabe
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Richard Fishel
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Ross C Larue
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
24
|
Johnson NM, Alvarado AF, Moffatt TN, Edavettal JM, Swaminathan TA, Braun SE. HIV-based lentiviral vectors: origin and sequence differences. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 21:451-465. [PMID: 33981779 PMCID: PMC8065252 DOI: 10.1016/j.omtm.2021.03.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 03/23/2021] [Indexed: 12/11/2022]
Abstract
Three gene therapy strategies have received US Food and Drug Administration (FDA) approval; one includes HIV-1-based lentiviral vectors. These vectors incorporate features to provide long-term gene transfer and expression while minimizing generation of a replication-competent virus or pathogenicity. Importantly, the coding regions of viral proteins were deleted, and the cis-acting regulatory elements were retained. With the use of representative vectors developed for clinical/commercial applications, we compared the vector backbone sequences to the initial sources of the HIV-1. All vectors included required elements: 5′ long terminal repeat (LTR) through the Ψ packaging signal, central polypurine tract/chain termination sequence (cPPT/CTS), Rev responsive element (RRE), and 3′ LTR, including a poly(A) signal. The Ψ signaling sequence demonstrated the greatest similarity between all vectors with only minor changes. The 3′ LTR was the most divergent sequence with a range of deletions. The RRE length varied between vectors. Phylogenetic analysis of the cPPT/CTS indicated multiple sources, perhaps because of its later inclusion into lentiviral vector systems, whereas other regions revealed node clusters around the HIV-1 reference genomes HXB2 and NL4-3. We examine the function of each region in a lentiviral vector, the molecular differences between vectors, and where optimization may guide development of the lentiviral delivery systems.
Collapse
Affiliation(s)
- Nathan M Johnson
- Division of Immunology, Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433, USA
| | - Anna Francesca Alvarado
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Trey N Moffatt
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Joshua M Edavettal
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Tarun A Swaminathan
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Stephen E Braun
- Division of Immunology, Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433, USA.,Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
25
|
Kuo CY, Kohn DB. Overview of the current status of gene therapy for primary immune deficiencies (PIDs). J Allergy Clin Immunol 2021; 146:229-233. [PMID: 32771134 DOI: 10.1016/j.jaci.2020.05.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/12/2020] [Accepted: 05/12/2020] [Indexed: 12/29/2022]
Abstract
Over 3 decades, gene therapy has advanced from a logical idea to becoming a clinical reality for several of the most severe primary immune deficiencies, as well as other inherited disorders. The first gene therapy medicines have been licensed for marketing and several more are advancing toward that goal to make them widely available, beyond clinical trials. Although common platforms of cells, vectors, or editing reagents are used for these disorders, each individual genetic cause of an immune deficiency requires its own vector or editing tools and a package of preclinical data on efficacy and safety to initiate clinical trials. One-by-one, gene therapy for primary immune deficiencies is being brought to the clinic and hopefully will provide safe and effective therapies.
Collapse
Affiliation(s)
- Caroline Y Kuo
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Calif
| | - Donald B Kohn
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Calif; Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Calif; Department of Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Calif; Department of Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research at UCLA, David Geffen School of Medicine, University of California, Los Angeles, Calif.
| |
Collapse
|
26
|
King JR, Notarangelo LD, Hammarström L. An appraisal of the Wilson & Jungner criteria in the context of genomic-based newborn screening for inborn errors of immunity. J Allergy Clin Immunol 2021; 147:428-438. [PMID: 33551024 PMCID: PMC8344044 DOI: 10.1016/j.jaci.2020.12.633] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/18/2020] [Accepted: 12/18/2020] [Indexed: 11/25/2022]
Abstract
Wilson and Jungner's recommendations for population-based screening have been used to guide decisions regarding candidate disease inclusion in newborn screening programs for the past 50 years. The advent of genomic-based technologies, including next-generation sequencing and its potential application to newborn screening, along with a changing landscape in terms of modern clinical practice and ethical, social, and legal considerations has led to a call for review of these criteria. Inborn errors of immunity (IEI) are a heterogeneous group of more than 450 genetically determined disorders of immunity, which are associated with significant morbidity and mortality, particularly where diagnosis and treatment are delayed. We argue that in addition to screening for severe combined immunodeficiency disease, which has already been initiated in several countries, other clinically significant IEI should be screened for at birth. Because of disease heterogeneity and identifiable genetic targets, a next-generation sequencing-based screening approach would be most suitable. A combination of worldwide experience and technological advances has improved our ability to diagnose and effectively treat patients with IEI. Considering IEI in the context of updated recommendations for population-based screening supports their potential inclusion as disease targets in newborn screening programs.
Collapse
Affiliation(s)
- Jovanka R King
- Department of Clinical Immunology, Karolinska University Hospital Huddinge, Stockholm, Sweden; Department of Immunopathology, SA Pathology, Women's and Children's Hospital Campus, Adelaide, Australia; Robinson Research Institute and Discipline of Paediatrics, School of Medicine, University of Adelaide, Adelaide, Australia
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Lennart Hammarström
- Department of Clinical Immunology, Karolinska University Hospital Huddinge, Stockholm, Sweden.
| |
Collapse
|
27
|
Liu L, Gao H, Guo C, Liu T, Li N, Qian Q. Therapeutic Mechanism of Nucleic Acid Drugs. ChemistrySelect 2021. [DOI: 10.1002/slct.202002901] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Lianxiao Liu
- Nucleic Acid Drug Division Shanghai Cell Therapy Group Co., Ltd. 75 A Qianyang Rd, Jiading District Shanghai 201805 China
| | - Haixia Gao
- Nucleic Acid Drug Division Shanghai Cell Therapy Group Co., Ltd. 75 A Qianyang Rd, Jiading District Shanghai 201805 China
| | - Chuanxin Guo
- Nucleic Acid Drug Division Shanghai Cell Therapy Group Co., Ltd. 75 A Qianyang Rd, Jiading District Shanghai 201805 China
| | - Tao Liu
- Nucleic Acid Drug Division Shanghai Cell Therapy Group Co., Ltd. 75 A Qianyang Rd, Jiading District Shanghai 201805 China
| | - Ning Li
- Nucleic Acid Drug Division Shanghai Cell Therapy Group Co., Ltd. 75 A Qianyang Rd, Jiading District Shanghai 201805 China
| | - Qijun Qian
- Nucleic Acid Drug Division Shanghai Cell Therapy Group Co., Ltd. 75 A Qianyang Rd, Jiading District Shanghai 201805 China
| |
Collapse
|
28
|
Molecular Imaging of Gene Therapy. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00064-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
29
|
Blanco E, Izotova N, Booth C, Thrasher AJ. Immune Reconstitution After Gene Therapy Approaches in Patients With X-Linked Severe Combined Immunodeficiency Disease. Front Immunol 2020; 11:608653. [PMID: 33329605 PMCID: PMC7729079 DOI: 10.3389/fimmu.2020.608653] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/02/2020] [Indexed: 12/21/2022] Open
Abstract
X-linked severe immunodeficiency disease (SCID-X1) is an inherited, rare, and life-threating disease. The genetic origin is a defect in the interleukin 2 receptor γ chain (IL2RG) gene and patients are classically characterized by absence of T and NK cells, as well as presence of partially-functional B cells. Without any treatment the disease is usually lethal during the first year of life. The treatment of choice for these patients is hematopoietic stem cell transplantation, with an excellent survival rate (>90%) if an HLA-matched sibling donor is available. However, when alternative donors are used, the success and survival rates are often lower. Gene therapy has been developed as an alternative treatment initially using γ-retroviral vectors to correct the defective γ chain in the absence of pre-conditioning treatment. The results were highly promising in SCID-X1 infants, showing long-term T-cell recovery and clinical benefit, although NK and B cell recovery was less robust. However, some infants developed T-cell acute lymphoblastic leukemia after the gene therapy, due to vector-mediated insertional mutagenesis. Consequently, considerable efforts have been made to develop safer vectors. The most recent clinical trials using lentiviral vectors together with a low-dose pre-conditioning regimen have demonstrated excellent sustained T cell recovery, but also B and NK cells, in both children and adults. This review provides an overview about the different gene therapy approaches used over the last 20 years to treat SCID-X1 patients, particularly focusing on lymphoid immune reconstitution, as well as the developments that have improved the process and outcomes.
Collapse
Affiliation(s)
- Elena Blanco
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Natalia Izotova
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Claire Booth
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
- Department of Paediatric Immunology, Great Ormond Street Hospital NHS Trust, London, United Kingdom
| | - Adrian James Thrasher
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
- Department of Paediatric Immunology, Great Ormond Street Hospital NHS Trust, London, United Kingdom
| |
Collapse
|
30
|
Canté-Barrett K, Staal FJT. An adequate human T cell repertoire from a single T cell progenitor: Lessons from an experiment of nature. EBioMedicine 2020; 60:103015. [PMID: 32977163 PMCID: PMC7516063 DOI: 10.1016/j.ebiom.2020.103015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 09/07/2020] [Indexed: 11/17/2022] Open
|
31
|
André I, Simons L, Ma K, Moirangthem RD, Diana JS, Magrin E, Couzin C, Magnani A, Cavazzana M. Ex vivo generated human T-lymphoid progenitors as a tool to accelerate immune reconstitution after partially HLA compatible hematopoietic stem cell transplantation or after gene therapy. Bone Marrow Transplant 2020; 54:749-755. [PMID: 31431705 DOI: 10.1038/s41409-019-0599-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Prolonged T-cell immunodeficiency following HLA- incompatible hematopoietic stem cell transplantation (HSCT) represents a major obstacle hampering the more widespread use of this approach. Strategies to fasten T-cell reconstitution in this setting are highly warranted as opportunistic infections and an increased risk of relapse account for high rates of morbidity and mortality especially during early month following this type of HSCT. We have implemented a feeder free cell system based on the use of the notch ligand DL4 and cytokines allowing for the in vitro differentiation of human T-Lymphoid Progenitor cells (HTLPs) from various sources of CD34+ hematopoietic stem and precursor cells (HSPCs). Co- transplantion of human T-lymphoid progenitors (HTLPs) and non- manipulated HSPCs into immunodeficient mice successfully accelerated the reconstitution of a polyclonal T-cell repertoire. This review summarizes preclinical data on the use of T-cell progenitors for treatment of post- transplantation immunodeficiency and gives insights into the development of GMP based protocols for potential clinical applications including gene therapy approaches. Future clinical trials implementing this protocol will aim at the acceleration of immune reconstitution in different clinical settings such as SCID and leukemia patients undergoing allogeneic transplantation. Apart from pure cell-therapy approaches, the combination of DL-4 culture with gene transduction protocols will open new perspectives in terms of gene therapy applications for primary immunodeficiencies.
Collapse
Affiliation(s)
- Isabelle André
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France. .,Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Imagine Institute, Paris, France. .,Paris Descartes University - Sorbonne Paris Cité, Imagine Institute, Paris, France.
| | - Laura Simons
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France.,Department of Biotherapy, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Kuiying Ma
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France.,Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Imagine Institute, Paris, France.,Paris Descartes University - Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - Ranjita Devi Moirangthem
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France.,Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Imagine Institute, Paris, France.,Paris Descartes University - Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - Jean-Sébastien Diana
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France.,Department of Biotherapy, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Elisa Magrin
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France.,Department of Biotherapy, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Chloé Couzin
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France.,Department of Biotherapy, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Alessandra Magnani
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France.,Department of Biotherapy, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Marina Cavazzana
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France.,Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Imagine Institute, Paris, France.,Paris Descartes University - Sorbonne Paris Cité, Imagine Institute, Paris, France.,Department of Biotherapy, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
32
|
Brommel CM, Cooney AL, Sinn PL. Adeno-Associated Virus-Based Gene Therapy for Lifelong Correction of Genetic Disease. Hum Gene Ther 2020; 31:985-995. [PMID: 32718227 PMCID: PMC7495917 DOI: 10.1089/hum.2020.138] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 07/27/2020] [Indexed: 12/27/2022] Open
Abstract
The list of successful gene therapy trials using adeno-associated virus (AAV)-based vectors continues to grow and includes a wide range of monogenic diseases. Replication incompetent AAV genomes typically remain episomal and expression dilutes as cells divide and die. Consequently, long-term transgene expression from AAV is best suited for quiescent cell types, such as retinal cells, myocytes, or neurons. For genetic diseases that involve cells with steady turnover, AAV-conferred correction may require routine readministration, where every dose carries the risk of developing an adaptive immune response that renders treatment ineffective. Here, we discuss innovative approaches to permanently modify the host genome using AAV-based platforms, thus potentially requiring only a single dose. Such approaches include using AAV delivery of DNA transposons, homologous recombination templates into safe harbors, and nucleases for targeting integration. In tissues with continual cell turnover, genetic modification of progenitor cell populations will help ensure persistent therapeutic outcomes. Combining the safety profile of AAV-based gene therapy vectors with the ability to integrate a therapeutic transgene creates novel solutions to the challenge of lifelong curative treatments for human genetic diseases.
Collapse
Affiliation(s)
| | - Ashley L. Cooney
- Department of Pediatrics, University of Iowa, Iowa City, Iowa, USA
| | - Patrick L. Sinn
- Program in Molecular Medicine, University of Iowa, Iowa City, Iowa, USA
- Department of Pediatrics, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
33
|
Garcia-Perez L, van Eggermond M, van Roon L, Vloemans SA, Cordes M, Schambach A, Rothe M, Berghuis D, Lagresle-Peyrou C, Cavazzana M, Zhang F, Thrasher AJ, Salvatori D, Meij P, Villa A, Van Dongen JJ, Zwaginga JJ, van der Burg M, Gaspar HB, Lankester A, Staal FJ, Pike-Overzet K. Successful Preclinical Development of Gene Therapy for Recombinase-Activating Gene-1-Deficient SCID. Mol Ther Methods Clin Dev 2020; 17:666-682. [PMID: 32322605 PMCID: PMC7163047 DOI: 10.1016/j.omtm.2020.03.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 03/23/2020] [Indexed: 12/19/2022]
Abstract
Recombinase-activating gene-1 (RAG1)-deficient severe combined immunodeficiency (SCID) patients lack B and T lymphocytes due to the inability to rearrange immunoglobulin and T cell receptor genes. Gene therapy is an alternative for those RAG1-SCID patients who lack a suitable bone marrow donor. We designed lentiviral vectors with different internal promoters driving codon-optimized RAG1 to ensure optimal expression. We used Rag1 -/- mice as a preclinical model for RAG1-SCID to assess the efficacy of the various vectors. We observed that B and T cell reconstitution directly correlated with RAG1 expression. Mice with low RAG1 expression showed poor immune reconstitution; however, higher expression resulted in phenotypic and functional lymphocyte reconstitution comparable to mice receiving wild-type stem cells. No signs of genotoxicity were found. Additionally, RAG1-SCID patient CD34+ cells transduced with our clinical RAG1 vector and transplanted into NSG mice led to improved human B and T cell development. Considering this efficacy outcome, together with favorable safety data, these results substantiate the need for a clinical trial for RAG1-SCID.
Collapse
Affiliation(s)
- Laura Garcia-Perez
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Marja van Eggermond
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Lieke van Roon
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Sandra A. Vloemans
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Martijn Cordes
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Michael Rothe
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Dagmar Berghuis
- Willem-Alexander Children’s Hospital Department of Pediatrics, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Chantal Lagresle-Peyrou
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France
- Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Imagine Institute and Paris Descartes University-Sorbonne Paris Cité, 75015 Paris, France
- Department of Biotherapy, Necker Children’s Hospital, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Marina Cavazzana
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France
- Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Imagine Institute and Paris Descartes University-Sorbonne Paris Cité, 75015 Paris, France
- Department of Biotherapy, Necker Children’s Hospital, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Fang Zhang
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, and Great Ormond Street Hospital NHS Trust, London WC1N 1EH, UK
| | - Adrian J. Thrasher
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, and Great Ormond Street Hospital NHS Trust, London WC1N 1EH, UK
| | - Daniela Salvatori
- Central Laboratory Animal Facility, Pathology Unit, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
- Department of Pharmacy, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
- Pathogenesis and Treatment of Immune and Bone Diseases Unit, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Anatomy and Physiology Division, Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan1, 3584CL Utrecht, the Netherlands
| | - Pauline Meij
- Department of Pharmacy, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Anna Villa
- Pathogenesis and Treatment of Immune and Bone Diseases Unit, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Jacques J.M. Van Dongen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Jaap-Jan Zwaginga
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Mirjam van der Burg
- Willem-Alexander Children’s Hospital Department of Pediatrics, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - H. Bobby Gaspar
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, and Great Ormond Street Hospital NHS Trust, London WC1N 1EH, UK
| | - Arjan Lankester
- Willem-Alexander Children’s Hospital Department of Pediatrics, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Frank J.T. Staal
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Karin Pike-Overzet
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| |
Collapse
|
34
|
Michniacki TF, Seth D, Secord E. Severe Combined Immunodeficiency: A Review for Neonatal Clinicians. Neoreviews 2020; 20:e326-e335. [PMID: 31261096 DOI: 10.1542/neo.20-6-e326] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The proper development and function of T cells is imperative in the creation of adequate cell-mediated and humoral immunity. Healthy term newborns have baseline immune immaturity, increasing their risk of infections, but significant immunologic consequences can occur, because of abnormal T-cell maturation. Combined immunodeficiencies can result, because B cells and natural killer cells rely on successful interactions with T cells to ensure their proper performance and survival. Severe combined immunodeficiency (SCID) is the most noteworthy of these conditions, leading to considerable early morbidity and often death by the age of 1 year if left untreated. Newborn screening for SCID is effective and allows for early implementation of lifesaving supportive measures, including protective isolation, initiation of prophylactic antimicrobials, caution with blood product transfusions, and avoidance of live vaccinations. Once a definitive diagnosis of SCID has been established, treatment frequently involves bone marrow or stem cell transplantation; however, enzyme replacement and gene therapy are also becoming options in those with SCID due to adenosine deaminase deficiency and other forms of SCID. Neonatal clinicians should understand the screening and diagnostic approach to SCID along with the initial management approaches for these extremely high-risk patients.
Collapse
Affiliation(s)
- Thomas F Michniacki
- Pediatrics and Communicable Diseases, Division of Pediatric Hematology/Oncology, University of Michigan, Ann Arbor, MI
| | - Divya Seth
- Department of Pediatrics, Division of Allergy, Asthma, & Immunology, Wayne State University, Detroit, MI
| | - Elizabeth Secord
- Department of Pediatrics, Division of Allergy, Asthma, & Immunology, Wayne State University, Detroit, MI
| |
Collapse
|
35
|
Lung P, Yang J, Li Q. Nanoparticle formulated vaccines: opportunities and challenges. NANOSCALE 2020; 12:5746-5763. [PMID: 32124894 DOI: 10.1039/c9nr08958f] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Vaccines harness the inherent properties of the immune system to prevent diseases or treat existing ones. Continuous efforts have been devoted to both gaining a mechanistic understanding of how the immune system operates and designing vaccines with high efficacies and effectiveness. Advancements in nanotechnology in recent years have generated unique opportunities to meet the daunting challenges associated with immunology and vaccine development. Firstly, nanoparticle formulated systems provide ideal model systems for studying the operation of the immune system, making it possible to systematically identify key factors and understand their roles in specific immune responses. Also, the versatile compositions/architectures of nanoparticle systems enable new strategies/novel platforms for developing vaccines with high efficacies and effectiveness. In this review, we discuss the advantages of nanoparticles and the challenges faced during vaccine development, through the framework of the immunological mechanisms of vaccination, with the aim of bridging the gap between immunology and materials science, which are both involved in vaccine design. The knowledge obtained provides general guidelines for future vaccine development.
Collapse
Affiliation(s)
- Pingsai Lung
- Department of Physics, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.
| | | | | |
Collapse
|
36
|
Weng Y, Li C, Yang T, Hu B, Zhang M, Guo S, Xiao H, Liang XJ, Huang Y. The challenge and prospect of mRNA therapeutics landscape. Biotechnol Adv 2020; 40:107534. [PMID: 32088327 DOI: 10.1016/j.biotechadv.2020.107534] [Citation(s) in RCA: 209] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 02/05/2020] [Accepted: 02/15/2020] [Indexed: 12/13/2022]
Abstract
Messenger RNA (mRNA)-based therapeutics hold the potential to cause a major revolution in the pharmaceutical industry because they can be used for precise and individualized therapy, and enable patients to produce therapeutic proteins in their own bodies without struggling with the comprehensive manufacturing issues associated with recombinant proteins. Compared with the current therapeutics, the production of mRNA is much cost-effective, faster and more flexible because it can be easily produced by in vitro transcription, and the process is independent of mRNA sequence. Moreover, mRNA vaccines allow people to develop personalized medications based on sequencing results and/or personalized conditions rapidly. Along with the great potential from bench to bedside, technical obstacles facing mRNA pharmaceuticals are also obvious. The stability, immunogenicity, translation efficiency, and delivery are all pivotal issues need to be addressed. In the recently published research results, these issues are gradually being overcome by state-of-the-art development technologies. In this review, we describe the structural properties and modification technologies of mRNA, summarize the latest advances in developing mRNA delivery systems, review the preclinical and clinical applications, and put forward our views on the prospect and challenges of developing mRNA into a new class of drug.
Collapse
Affiliation(s)
- Yuhua Weng
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, PR China
| | - Chunhui Li
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, PR China
| | - Tongren Yang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, PR China
| | - Bo Hu
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, PR China
| | - Mengjie Zhang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, PR China
| | - Shuai Guo
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, PR China
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Xing-Jie Liang
- Chinese Academy of Sciences (CAS), Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, PR China
| | - Yuanyu Huang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, PR China.
| |
Collapse
|
37
|
Brokowski C, Adli M. Ethical Considerations in Therapeutic Clinical Trials Involving Novel Human Germline-Editing Technology. CRISPR J 2020; 3:18-26. [DOI: 10.1089/crispr.2019.0051] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Carolyn Brokowski
- Department of Emergency Medicine, Yale School of Medicine, New Haven, Connecticut; and Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern, Chicago, Illinois
| | - Mazhar Adli
- Robert Lurie Comprehensive Cancer Center, Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern, Chicago, Illinois
| |
Collapse
|
38
|
Nochi T, Suzuki S, Ito S, Morita S, Furukawa M, Fuchimoto D, Sasahara Y, Usami K, Niimi K, Itano O, Kitago M, Matsuda S, Matsuo A, Suyama Y, Sakai Y, Wu G, Bazer FW, Watanabe K, Onishi A, Aso H. Elucidation of the Effects of a Current X-SCID Therapy on Intestinal Lymphoid Organogenesis Using an In Vivo Animal Model. Cell Mol Gastroenterol Hepatol 2020; 10:83-100. [PMID: 32017983 PMCID: PMC7210612 DOI: 10.1016/j.jcmgh.2020.01.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 01/26/2020] [Accepted: 01/27/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND & AIMS Organ-level research using an animal model lacking Il2rg, the gene responsible for X-linked severe combined immunodeficiency (X-SCID), is clinically unavailable and would be a powerful tool to gain deeper insights into the symptoms of patients with X-SCID. METHODS We used an X-SCID animal model, which was first established in our group by the deletion of Il2rg gene in pigs, to understand the clinical signs from multiple perspectives based on pathology, immunology, microbiology, and nutrition. We also treated the X-SCID pigs with bone marrow transplantation (BMT) for mimicking a current therapeutic treatment for patients with X-SCID and investigated the effect at the organ-level. Moreover, the results were confirmed using serum and fecal samples collected from patients with X-SCID. RESULTS We demonstrated that X-SCID pigs completely lacked Peyer's patches (PPs) and IgA production in the small intestine, but possessed some dysfunctional intestinal T and B cells. Another novel discovery was that X-SCID pigs developed a heterogeneous intestinal microflora and possessed abnormal plasma metabolites, indicating that X-SCID could be an immune disorder that affects various in vivo functions. Importantly, the organogenesis of PPs in X-SCID pigs was not promoted by BMT. Although a few isolated lymphoid follicles developed in the small intestine of BMT-treated X-SCID pigs, there was no evidence that they contributed to IgA production and microflora formation. Consistently, most patients with X-SCID who received BMT possessed abnormal intestinal immune and microbial environments regardless of the presence of sufficient serum IgG. CONCLUSIONS These results indicate that the current BMT therapies for patients with X-SCID may be insufficient to induce the organogenesis of intestinal lymphoid tissues that are associated with numerous functions in vivo.
Collapse
Affiliation(s)
- Tomonori Nochi
- International Education and Research Center for Food and Agricultural Immunology, Tohoku University Graduate School of Agricultural Science, Miyagi, Japan,International Research and Development Center for Mucosal Vaccine, Institute of Medical Science, University of Tokyo, Tokyo, Japan,Correspondence Address correspondence to: Tomonori Nochi, International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, 468-1 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8572, Japan. fax: +81-22-757-4315.
| | - Shunichi Suzuki
- Division of Animal Science, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Japan
| | - Shun Ito
- International Education and Research Center for Food and Agricultural Immunology, Tohoku University Graduate School of Agricultural Science, Miyagi, Japan
| | - Shotaro Morita
- International Education and Research Center for Food and Agricultural Immunology, Tohoku University Graduate School of Agricultural Science, Miyagi, Japan
| | - Mutsumi Furukawa
- International Education and Research Center for Food and Agricultural Immunology, Tohoku University Graduate School of Agricultural Science, Miyagi, Japan
| | - Daiichiro Fuchimoto
- Division of Animal Science, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Japan
| | - Yoji Sasahara
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Katsuki Usami
- International Education and Research Center for Food and Agricultural Immunology, Tohoku University Graduate School of Agricultural Science, Miyagi, Japan
| | - Kanae Niimi
- International Education and Research Center for Food and Agricultural Immunology, Tohoku University Graduate School of Agricultural Science, Miyagi, Japan
| | - Osamu Itano
- Department of Hepato-Biliary-Pancreatic and Gastrointestinal Surgery, International University of Health and Welfare School of Medicine, Chiba, Japan
| | - Minoru Kitago
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Sachiko Matsuda
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Ayumi Matsuo
- International Education and Research Center for Food and Agricultural Immunology, Tohoku University Graduate School of Agricultural Science, Miyagi, Japan
| | - Yoshihisa Suyama
- International Education and Research Center for Food and Agricultural Immunology, Tohoku University Graduate School of Agricultural Science, Miyagi, Japan
| | - Yoshifumi Sakai
- International Education and Research Center for Food and Agricultural Immunology, Tohoku University Graduate School of Agricultural Science, Miyagi, Japan
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, Texas
| | - Fuller W. Bazer
- Department of Animal Science, Texas A&M University, College Station, Texas
| | - Kouichi Watanabe
- International Education and Research Center for Food and Agricultural Immunology, Tohoku University Graduate School of Agricultural Science, Miyagi, Japan
| | - Akira Onishi
- Department of Animal Science and Resources, Nihon University College of Bioresource Sciences, Kanagawa, Japan
| | - Hisashi Aso
- International Education and Research Center for Food and Agricultural Immunology, Tohoku University Graduate School of Agricultural Science, Miyagi, Japan
| |
Collapse
|
39
|
Intact Viral Particle Counts Measured by Flow Virometry Provide Insight into the Infectivity and Genome Packaging Efficiency of Moloney Murine Leukemia Virus. J Virol 2020; 94:JVI.01600-19. [PMID: 31694951 DOI: 10.1128/jvi.01600-19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 10/22/2019] [Indexed: 02/06/2023] Open
Abstract
Murine leukemia viruses (MLVs) have long been used as a research model to further our understanding of retroviruses. These simple gammaretroviruses have been studied extensively in various facets of science for nearly half a century, yet we have surprisingly little quantitative information about some of the basic features of these viral particles. These include parameters such as the genome packaging efficiency and the number of particles required for a productive infection. The reason for this knowledge gap relies primarily on the technical challenge of accurately measuring intact viral particles from infected cell supernatants. Virus-infected cells are well known to release soluble viral proteins, defective viruses, and extracellular vesicles (EVs) harboring viral proteins that may mimic viruses, all of which can skew virus titer quantifications. Flow virometry, also known as nanoscale flow cytometry or simply small-particle flow cytometry, is an emerging analytical method enabling high-throughput single-virus phenotypic characterizations. By utilizing the viral envelope glycoprotein (Env) and monodisperse light scattering characteristics as discerning parameters of intact virus particles, here, we analyzed the basic properties of Moloney MLV (M-MLV). We show that <24% of the total p30 capsid protein measured in infected cell supernatants is associated with intact viruses. We calculate that about one in five M-MLV particles contains a viral RNA genome pair and that individual intact particle infectivity is about 0.4%. These findings provide new insights into the characteristics of an extensively studied prototypical retrovirus while highlighting the benefits of flow virometry for the field of virology.IMPORTANCE Gammaretroviruses, or, more specifically, murine leukemia viruses (MLVs), have been a longstanding model for studying retroviruses. Although being extensively analyzed and dissected for decades, several facets of MLV biology are still poorly understood. One of the primary challenges has been enumerating total intact virus particles in a sample. While several analytical methods can precisely measure virus protein amounts, MLVs are known to induce the secretion of soluble and vesicle-associated viral proteins that can skew these measurements. With recent technological advances in flow cytometry, it is now possible to analyze viruses down to 90 nm in diameter with an approach called flow virometry. The technique has the added benefit of being able to discriminate viruses from extracellular vesicles and free viral proteins in order to confidently provide an intact viral particle count. Here, we used flow virometry to provide new insights into the basic characteristics of Moloney MLV.
Collapse
|
40
|
Miah KM, Hyde SC, Gill DR. Emerging gene therapies for cystic fibrosis. Expert Rev Respir Med 2019; 13:709-725. [PMID: 31215818 DOI: 10.1080/17476348.2019.1634547] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 06/18/2019] [Indexed: 01/06/2023]
Abstract
Introduction: Cystic fibrosis (CF) remains a life-threatening genetic disease, with few clinically effective treatment options. Gene therapy and gene editing strategies offer the potential for a one-time CF cure, irrespective of the CFTR mutation class. Areas covered: We review emerging gene therapies and gene delivery strategies for the treatment of CF particularly viral and non-viral approaches with potential to treat CF. Expert opinion: It was initially anticipated that the challenge of developing a gene therapy for CF lung disease would be met relatively easily. Following early proof-of-concept clinical studies, CF gene therapy has entered a new era with innovative vector designs, approaches to subvert the humoral immune system and increase gene delivery and gene correction efficiencies. Developments include integrating adenoviral vectors, rapamycin-loaded nanoparticles, and lung-tropic lentiviral vectors. The characterization of novel cell types in the lung epithelium, including pulmonary ionocytes, may also encourage cell type-specific targeting for CF correction. We anticipate preclinical studies to further validate these strategies, which should pave the way for clinical trials. We also expect gene editing efficiencies to improve to clinically translatable levels, given advancements in viral and non-viral vectors. Overall, gene delivery technologies look more convincing in producing an effective CF gene therapy.
Collapse
Affiliation(s)
- Kamran M Miah
- a Gene Medicine Group, Nuffield Division of Clinical Laboratory Science, Radcliffe Department of Medicine, University of Oxford , Oxford , UK
| | - Stephen C Hyde
- a Gene Medicine Group, Nuffield Division of Clinical Laboratory Science, Radcliffe Department of Medicine, University of Oxford , Oxford , UK
| | - Deborah R Gill
- a Gene Medicine Group, Nuffield Division of Clinical Laboratory Science, Radcliffe Department of Medicine, University of Oxford , Oxford , UK
| |
Collapse
|
41
|
Arabipour I, Amani J, Mirhosseini SA, Salimian J. The study of genes and signal transduction pathways involved in mustard lung injury: A gene therapy approach. Gene 2019; 714:143968. [PMID: 31323308 DOI: 10.1016/j.gene.2019.143968] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 07/06/2019] [Accepted: 07/08/2019] [Indexed: 02/06/2023]
Abstract
Sulfur mustard (SM) is a destructive and harmful chemical agent for the eyes, skin and lungs that causes short-term and long-term lesions and was widely used in Iraq war against Iran (1980-1988). SM causes DNA damages, oxidative stress, and Inflammation. Considering the similarities between SM and COPD (Chronic Obstructive Pulmonary Disease) pathogens and limited available treatments, a novel therapeutic approach is not developed. Gene therapy is a novel therapeutic approach that uses genetic engineering science in treatment of most diseases including chronic obstructive pulmonary disease. In this review, attempts to presenting a comprehensive study of mustard lung and introducing the genes therapy involved in chronic obstructive pulmonary disease and emphasizing the pathways and genes involved in the pathology and pathogenesis of sulfur Mustard. It seems that, given the high potential of gene therapy and the fact that this experimental technique is a candidate for the treatment of pulmonary diseases, further study of genes, vectors and gene transfer systems can draw a very positive perspective of gene therapy in near future.
Collapse
Affiliation(s)
- Iman Arabipour
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Jafar Amani
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Seyed Ali Mirhosseini
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Jafar Salimian
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
42
|
Ghani K, Boivin-Welch M, Roy S, Dakiw-Piaceski A, Barbier M, Pope E, Germain L, Caruso M. Generation of High-Titer Self-Inactivated γ-Retroviral Vector Producer Cells. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 14:90-99. [PMID: 31312667 PMCID: PMC6610700 DOI: 10.1016/j.omtm.2019.05.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 05/30/2019] [Indexed: 12/01/2022]
Abstract
The γ-retroviral vector is a gene delivery vehicle that is commonly used in gene therapy. Despite its efficacy, its strong enhancers contributed to malignant transformations in some hematopoietic stem cell (HSC) gene therapy trials. A safer version without viral enhancers (SIN) is available, but its production is cumbersome, as high titers can only be obtained in transient transfection. Our aim was to develop a system that could easily generate high-titer SIN vectors from stable producer cells. The use of the cytomegalovirus enhancer-promoter sequence to generate the full-length genomic RNA combined to sequences that decrease transcriptional readthrough (WPRE and strong polyadenylation sequences) led to 6 × 106 infectious units (IU)/mL of a SIN GFP vector in transient transfection. The incorporation of a blasticidin selection cassette to the retroviral plasmid allowed the generation of stable clones in the 293Vec packaging cells that release 2 × 107 IU/mL and 1.4 × 107 IU/mL of a SIN GFP and a SIN PIGA vector, respectively. A titer of 1.8 × 106 IU/mL was obtained with a SIN vector containing the long 8.9-kb COL7A1 cDNA. Thus, an efficient process was established for the generation of stable 293Vec-derived retrovirus producer cells that release high-titer SIN vectors.
Collapse
Affiliation(s)
- Karim Ghani
- CHU de Québec-Université Laval Research Center (Oncology Division), Université Laval Cancer Research Center, and Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Québec, QC G1R 2J6, Canada
| | - Michael Boivin-Welch
- CHU de Québec-Université Laval Research Center (Oncology Division), Université Laval Cancer Research Center, and Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Québec, QC G1R 2J6, Canada.,CHU de Québec-Université Laval Research Center (Regenerative Medicine Division) and Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, and Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, G1J 1Z4, Canada
| | - Sylvie Roy
- CHU de Québec-Université Laval Research Center (Oncology Division), Université Laval Cancer Research Center, and Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Québec, QC G1R 2J6, Canada
| | - Angela Dakiw-Piaceski
- CHU de Québec-Université Laval Research Center (Regenerative Medicine Division) and Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, and Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, G1J 1Z4, Canada
| | - Martin Barbier
- CHU de Québec-Université Laval Research Center (Regenerative Medicine Division) and Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, and Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, G1J 1Z4, Canada
| | - Elena Pope
- Section of Dermatology, The Hospital for Sick Children and University of Toronto, Toronto, ON, Canada
| | - Lucie Germain
- CHU de Québec-Université Laval Research Center (Regenerative Medicine Division) and Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, and Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, G1J 1Z4, Canada
| | - Manuel Caruso
- CHU de Québec-Université Laval Research Center (Oncology Division), Université Laval Cancer Research Center, and Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Québec, QC G1R 2J6, Canada
| |
Collapse
|
43
|
Peptide-mediated delivery of therapeutic mRNA in ovarian cancer. Eur J Pharm Biopharm 2019; 141:180-190. [PMID: 31103743 DOI: 10.1016/j.ejpb.2019.05.014] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/13/2019] [Accepted: 05/15/2019] [Indexed: 12/27/2022]
Abstract
Ovarian cancer is the most lethal gynecological malignancy in the developed world. In spite of intensive research, the mortality has hardly decreased over the past twenty years. This necessitates the exploration of novel therapeutic modalities. Transient protein expression through delivery of mRNA is emerging as a highly promising option. In contrast to gene therapy there is no risk of integration into the genome. Here, we explore the expression of mRNA in models of ovarian cancer of increasing complexity. The cell-penetrating peptide (CPP) PepFect 14 (PF14) was used to formulate CPP-mRNA nanoparticles. Efficient expression of a reporter protein was achieved in two-dimensional tissue cultures and in three-dimensional cancer cell spheroids. PF14 nanoparticles greatly outperformed a lipid-based transfection agent in vivo, leading to expression in various cell types of tumor associated tissue. Protein expression was restricted to the peritoneal cavity. Messenger RNA expression across different cell types was confirmed in primary ovarian cancer explants. As ovarian cancer is confined to the peritoneal cavity in most cases, the results create the basis for applications in which the tumor microenvironment is transiently modified through protein expression.
Collapse
|
44
|
Pavel-Dinu M, Wiebking V, Dejene BT, Srifa W, Mantri S, Nicolas CE, Lee C, Bao G, Kildebeck EJ, Punjya N, Sindhu C, Inlay MA, Saxena N, DeRavin SS, Malech H, Roncarolo MG, Weinberg KI, Porteus MH. Gene correction for SCID-X1 in long-term hematopoietic stem cells. Nat Commun 2019; 10:1634. [PMID: 30967552 PMCID: PMC6456568 DOI: 10.1038/s41467-019-09614-y] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 03/12/2019] [Indexed: 12/28/2022] Open
Abstract
Gene correction in human long-term hematopoietic stem cells (LT-HSCs) could be an effective therapy for monogenic diseases of the blood and immune system. Here we describe an approach for X-linked sSevere cCombined iImmunodeficiency (SCID-X1) using targeted integration of a cDNA into the endogenous start codon to functionally correct disease-causing mutations throughout the gene. Using a CRISPR-Cas9/AAV6 based strategy, we achieve up to 20% targeted integration frequencies in LT-HSCs. As measures of the lack of toxicity we observe no evidence of abnormal hematopoiesis following transplantation and no evidence of off-target mutations using a high-fidelity Cas9 as a ribonucleoprotein complex. We achieve high levels of targeting frequencies (median 45%) in CD34+ HSPCs from six SCID-X1 patients and demonstrate rescue of lymphopoietic defect in a patient derived HSPC population in vitro and in vivo. In sum, our study provides specificity, toxicity and efficacy data supportive of clinical development of genome editing to treat SCID-Xl.
Collapse
Affiliation(s)
- Mara Pavel-Dinu
- Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Volker Wiebking
- Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Beruh T Dejene
- Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Waracharee Srifa
- Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Sruthi Mantri
- Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Carmencita E Nicolas
- Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Ciaran Lee
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Gang Bao
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Eric J Kildebeck
- Center for Engineering Innovation, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Niraj Punjya
- Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA
- University of California Davis, School of Medicine, Sacramento, CA, 95817, USA
| | - Camille Sindhu
- Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Matthew A Inlay
- Department of Cellular and Molecular Biosciences, University of California Irvine, Irvine, CA, 92697, USA
| | - Nivedita Saxena
- Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Suk See DeRavin
- Laboratory of Host Defenses, National Institutes of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD, 20892, USA
| | - Harry Malech
- Laboratory of Host Defenses, National Institutes of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD, 20892, USA
| | - Maria Grazia Roncarolo
- Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Kenneth I Weinberg
- Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Matthew H Porteus
- Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
45
|
Marquez Loza LI, Yuen EC, McCray PB. Lentiviral Vectors for the Treatment and Prevention of Cystic Fibrosis Lung Disease. Genes (Basel) 2019; 10:genes10030218. [PMID: 30875857 PMCID: PMC6471883 DOI: 10.3390/genes10030218] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 03/08/2019] [Accepted: 03/11/2019] [Indexed: 01/04/2023] Open
Abstract
Despite the continued development of cystic fibrosis transmembrane conductance regulator (CFTR) modulator drugs for the treatment of cystic fibrosis (CF), the need for mutation agnostic treatments remains. In a sub-group of CF individuals with mutations that may not respond to modulators, such as those with nonsense mutations, CFTR gene transfer to airway epithelia offers the potential for an effective treatment. Lentiviral vectors are well-suited for this purpose because they transduce nondividing cells, and provide long-term transgene expression. Studies in primary cultures of human CF airway epithelia and CF animal models demonstrate the long-term correction of CF phenotypes and low immunogenicity using lentiviral vectors. Further development of CF gene therapy requires the investigation of optimal CFTR expression in the airways. Lentiviral vectors with improved safety features have minimized insertional mutagenesis safety concerns raised in early clinical trials for severe combined immunodeficiency using γ-retroviral vectors. Recent clinical trials using improved lentiviral vectors support the feasibility and safety of lentiviral gene therapy for monogenetic diseases. While work remains to be done before CF gene therapy reaches the bedside, recent advances in lentiviral vector development reviewed here are encouraging and suggest it could be tested in clinical studies in the near future.
Collapse
Affiliation(s)
- Laura I Marquez Loza
- Stead Family Department of Pediatrics, The University of Iowa, Iowa City, IA 52242, USA.
- Pappajohn Biomedical Institute and the Center for Gene Therapy, The University of Iowa, Iowa City, IA 52242, USA.
| | - Eric C Yuen
- Talee Bio, 3001 Market Street, Suite 140, Philadelphia, PA 19104, USA.
| | - Paul B McCray
- Stead Family Department of Pediatrics, The University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
46
|
Gene therapy targeting haematopoietic stem cells for inherited diseases: progress and challenges. Nat Rev Drug Discov 2019; 18:447-462. [DOI: 10.1038/s41573-019-0020-9] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
47
|
Tajer P, Pike-Overzet K, Arias S, Havenga M, Staal FJT. Ex Vivo Expansion of Hematopoietic Stem Cells for Therapeutic Purposes: Lessons from Development and the Niche. Cells 2019; 8:cells8020169. [PMID: 30781676 PMCID: PMC6407064 DOI: 10.3390/cells8020169] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 02/08/2019] [Accepted: 02/13/2019] [Indexed: 12/21/2022] Open
Abstract
Expansion of hematopoietic stem cells (HSCs) for therapeutic purposes has been a “holy grail” in the field for many years. Ex vivo expansion of HSCs can help to overcome material shortage for transplantation purposes and genetic modification protocols. In this review, we summarize improved understanding in blood development, the effect of niche and conservative signaling pathways on HSCs in mice and humans, and also advances in ex vivo culturing protocols of human HSCs with cytokines or small molecule compounds. Different expansion protocols have been tested in clinical trials. However, an optimal condition for ex vivo expansion of human HSCs still has not been found yet. Translating and implementing new findings from basic research (for instance by using genetic modification of human HSCs) into clinical protocols is crucial to improve ex vivo expansion and eventually boost stem cell gene therapy.
Collapse
Affiliation(s)
- Parisa Tajer
- Department of Immunohematology and Blood Transfusion, L3-Q Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
- Department of Molecular Cell Biology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| | - Karin Pike-Overzet
- Department of Immunohematology and Blood Transfusion, L3-Q Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
- Department of Molecular Cell Biology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| | - Sagrario Arias
- Batavia Biosciences, Zernikedreef 16, 2333 CL Leiden, The Netherlands.
| | - Menzo Havenga
- Batavia Biosciences, Zernikedreef 16, 2333 CL Leiden, The Netherlands.
| | - Frank J T Staal
- Department of Immunohematology and Blood Transfusion, L3-Q Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
- Department of Molecular Cell Biology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| |
Collapse
|
48
|
Lino CA, Harper JC, Carney JP, Timlin JA. Delivering CRISPR: a review of the challenges and approaches. Drug Deliv 2018; 25:1234-1257. [PMID: 29801422 PMCID: PMC6058482 DOI: 10.1080/10717544.2018.1474964] [Citation(s) in RCA: 669] [Impact Index Per Article: 95.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 05/03/2018] [Accepted: 05/07/2018] [Indexed: 12/13/2022] Open
Abstract
Gene therapy has long held promise to correct a variety of human diseases and defects. Discovery of the Clustered Regularly-Interspaced Short Palindromic Repeats (CRISPR), the mechanism of the CRISPR-based prokaryotic adaptive immune system (CRISPR-associated system, Cas), and its repurposing into a potent gene editing tool has revolutionized the field of molecular biology and generated excitement for new and improved gene therapies. Additionally, the simplicity and flexibility of the CRISPR/Cas9 site-specific nuclease system has led to its widespread use in many biological research areas including development of model cell lines, discovering mechanisms of disease, identifying disease targets, development of transgene animals and plants, and transcriptional modulation. In this review, we present the brief history and basic mechanisms of the CRISPR/Cas9 system and its predecessors (ZFNs and TALENs), lessons learned from past human gene therapy efforts, and recent modifications of CRISPR/Cas9 to provide functions beyond gene editing. We introduce several factors that influence CRISPR/Cas9 efficacy which must be addressed before effective in vivo human gene therapy can be realized. The focus then turns to the most difficult barrier to potential in vivo use of CRISPR/Cas9, delivery. We detail the various cargos and delivery vehicles reported for CRISPR/Cas9, including physical delivery methods (e.g. microinjection; electroporation), viral delivery methods (e.g. adeno-associated virus (AAV); full-sized adenovirus and lentivirus), and non-viral delivery methods (e.g. liposomes; polyplexes; gold particles), and discuss their relative merits. We also examine several technologies that, while not currently reported for CRISPR/Cas9 delivery, appear to have promise in this field. The therapeutic potential of CRISPR/Cas9 is vast and will only increase as the technology and its delivery improves.
Collapse
Affiliation(s)
- Christopher A. Lino
- Bioenergy and Defense Technologies, Sandia National Laboratories, Albuquerque, NM, USA
| | - Jason C. Harper
- Bioenergy and Defense Technologies, Sandia National Laboratories, Albuquerque, NM, USA
| | - James P. Carney
- Bioenergy and Defense Technologies, Sandia National Laboratories, Albuquerque, NM, USA
| | - Jerilyn A. Timlin
- Bioenergy and Defense Technologies, Sandia National Laboratories, Albuquerque, NM, USA
| |
Collapse
|
49
|
Doshi BS, Arruda VR. Gene therapy for hemophilia: what does the future hold? Ther Adv Hematol 2018; 9:273-293. [PMID: 30210756 DOI: 10.1177/2040620718791933] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 07/09/2018] [Indexed: 01/19/2023] Open
Abstract
Recent phase I/II adeno-associated viral vector-mediated gene therapy clinical trials have reported remarkable success in ameliorating disease phenotype in hemophilia A and B. These trials, which highlight the challenges overcome through decades of preclinical and first in human clinical studies, have generated considerable excitement for patients and caregivers alike. Optimization of vector and transgene expression has significantly improved the ability to achieve therapeutic factor levels in these subjects. Long-term follow-up studies will guide standardization of the approach with respect to the combination of serotype, promoter, dose, and manufacturing processes and inform safety for inclusion of young patients. Certain limitations preclude universal applicability of gene therapy, including transient liver transaminase elevations due to the immune responses to vector capsids or as yet undefined mechanisms, underlying liver disease from iatrogenic viral hepatitis, and neutralizing antibodies to clotting factors. Integrating vectors show promising preclinical results, but manufacturing and safety concerns still remain. The prospect of gene editing for correction of the underlying mutation is on the horizon with considerable potential. Herein, we review the advances and limitations that have resulted in these recent successful clinical trials and outline avenues that will allow for broader applicability of gene therapy.
Collapse
Affiliation(s)
- Bhavya S Doshi
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Valder R Arruda
- Department of Pediatrics, The Children's Hospital of Philadelphia, 3501 Civic Center Blvd, 5056 Colket Translational Research Center, Philadelphia, PA 19104, USA
| |
Collapse
|
50
|
Ohmori T, Mizukami H, Katakai Y, Kawai S, Nakamura H, Inoue M, Shu T, Sugimoto H, Sakata Y. Safety of intra-articular transplantation of lentivirally transduced mesenchymal stromal cells for haemophilic arthropathy in a non-human primate. Int J Hematol 2018; 108:239-245. [PMID: 29737459 DOI: 10.1007/s12185-018-2465-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 04/25/2018] [Accepted: 04/26/2018] [Indexed: 12/01/2022]
Abstract
Joint bleeding and resultant arthropathy are major determinants of quality of life in haemophilia patients. We previously developed a mesenchymal stromal cell (MSC)-based treatment approach for haemophilic arthropathy in a mouse model of haemophilia A. Here, we evaluated the long-term safety of intra-articular injection of lentivirally transduced autologous MSCs in non-human primates. Autologous bone-marrow-derived MSCs transduced with a lentiviral vector expressing coagulation factor VIII (FVIII) were injected into the left knee joint of cynomolgus monkeys. We first conducted codon optimization to increase FVIII production in the cells. Lentiviral transduction of autologous MSCs resulted in a significant increase of FVIII in the culture supernatant before transplantation. We did not find any tumour generation around the knee structure at 11-16 months after injection by magnetic resonance imaging. The proviral sequence of the simian immunodeficiency virus lentiviral vector was not detected in the heart, lungs, spleen, liver, testis, or bone marrow by real-time quantitative PCR. We confirmed the long-term safety of intra-articular injection of transduced MSCs in a non-human primate. The procedure may be an attractive therapeutic approach for joint diseases in haemophilia patients.
Collapse
Affiliation(s)
- Tsukasa Ohmori
- Department of Biochemistry, Jichi Medical University School of Medicine, 3111-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan.
| | - Hiroaki Mizukami
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Yuko Katakai
- The Corporation for Production and Research of Laboratory Primates, 1-16-2 Sakura, Tsukuba, Ibaraki, 305-0003, Japan
| | - Sho Kawai
- Department of Radiology, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Hitoyasu Nakamura
- Department of Radiology, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Makoto Inoue
- ID Pharma Inc., 6 Okubo, Tsukuba, Ibaraki, 300-2611, Japan
| | - Tsugumine Shu
- ID Pharma Inc., 6 Okubo, Tsukuba, Ibaraki, 300-2611, Japan
| | - Hideharu Sugimoto
- Department of Radiology, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Yoichi Sakata
- Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| |
Collapse
|