1
|
Lemmens M, Dorsheimer L, Zeller A, Dietz-Baum Y. Non-clinical safety assessment of novel drug modalities: Genome safety perspectives on viral-, nuclease- and nucleotide-based gene therapies. MUTATION RESEARCH. GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2024; 896:503767. [PMID: 38821669 DOI: 10.1016/j.mrgentox.2024.503767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/08/2024] [Accepted: 05/13/2024] [Indexed: 06/02/2024]
Abstract
Gene therapies have emerged as promising treatments for various conditions including inherited diseases as well as cancer. Ensuring their safe clinical application requires the development of appropriate safety testing strategies. Several guidelines have been provided by health authorities to address these concerns. These guidelines state that non-clinical testing should be carried out on a case-by-case basis depending on the modality. This review focuses on the genome safety assessment of frequently used gene therapy modalities, namely Adeno Associated Viruses (AAVs), Lentiviruses, designer nucleases and mRNAs. Important safety considerations for these modalities, amongst others, are vector integrations into the patient genome (insertional mutagenesis) and off-target editing. Taking into account the constraints of in vivo studies, health authorities endorse the development of novel approach methodologies (NAMs), which are innovative in vitro strategies for genotoxicity testing. This review provides an overview of NAMs applied to viral and CRISPR/Cas9 safety, including next generation sequencing-based methods for integration site analysis and off-target editing. Additionally, NAMs to evaluate the oncogenicity risk arising from unwanted genomic modifications are discussed. Thus, a range of promising techniques are available to support the safe development of gene therapies. Thorough validation, comparisons and correlations with clinical outcomes are essential to identify the most reliable safety testing strategies. By providing a comprehensive overview of these NAMs, this review aims to contribute to a better understanding of the genome safety perspectives of gene therapies.
Collapse
Affiliation(s)
| | - Lena Dorsheimer
- Research and Development, Preclinical Safety, Sanofi, Industriepark Hoechst, Frankfurt am Main 65926, Germany.
| | - Andreas Zeller
- Pharmaceutical Sciences, pRED Innovation Center Basel, Hoffmann-La Roche Ltd, Basel 4070, Switzerland
| | - Yasmin Dietz-Baum
- Research and Development, Preclinical Safety, Sanofi, Industriepark Hoechst, Frankfurt am Main 65926, Germany
| |
Collapse
|
2
|
Sabatino DE, Bushman FD, Chandler RJ, Crystal RG, Davidson BL, Dolmetsch R, Eggan KC, Gao G, Gil-Farina I, Kay MA, McCarty DM, Montini E, Ndu A, Yuan J. Evaluating the state of the science for adeno-associated virus integration: An integrated perspective. Mol Ther 2022; 30:2646-2663. [PMID: 35690906 PMCID: PMC9372310 DOI: 10.1016/j.ymthe.2022.06.004] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/03/2022] [Accepted: 06/06/2022] [Indexed: 12/12/2022] Open
Abstract
On August 18, 2021, the American Society of Gene and Cell Therapy (ASGCT) hosted a virtual roundtable on adeno-associated virus (AAV) integration, featuring leading experts in preclinical and clinical AAV gene therapy, to further contextualize and understand this phenomenon. Recombinant AAV (rAAV) vectors are used to develop therapies for many conditions given their ability to transduce multiple cell types, resulting in long-term expression of transgenes. Although most rAAV DNA typically remains episomal, some rAAV DNA becomes integrated into genomic DNA at a low frequency, and rAAV insertional mutagenesis has been shown to lead to tumorigenesis in neonatal mice. Currently, the risk of rAAV-mediated oncogenesis in humans is theoretical because no confirmed genotoxic events have been reported to date. However, because insertional mutagenesis has been reported in a small number of murine studies, there is a need to characterize this genotoxicity to inform research, regulatory needs, and patient care. The purpose of this white paper is to review the evidence of rAAV-related host genome integration in animal models and possible risks of insertional mutagenesis in patients. In addition, technical considerations, regulatory guidance, and bioethics are discussed.
Collapse
Affiliation(s)
- Denise E Sabatino
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA; Division of Hematology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Frederic D Bushman
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Randy J Chandler
- National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - Ronald G Crystal
- Department of Genetic Medicine, Weill Medical College of Cornell University, New York, NY, USA
| | - Beverly L Davidson
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | | | - Mark A Kay
- Departments of Pediatrics and Genetics, Stanford University, Stanford, CA, USA
| | | | - Eugenio Montini
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Adora Ndu
- BridgeBio Pharma, Inc., Palo Alto, CA, USA
| | - Jing Yuan
- Drug Safety Research and Development, Pfizer Inc., Cambridge, MA, USA
| |
Collapse
|
3
|
Morgan MA, Galla M, Grez M, Fehse B, Schambach A. Retroviral gene therapy in Germany with a view on previous experience and future perspectives. Gene Ther 2021; 28:494-512. [PMID: 33753908 PMCID: PMC8455336 DOI: 10.1038/s41434-021-00237-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 01/13/2021] [Accepted: 02/01/2021] [Indexed: 02/01/2023]
Abstract
Gene therapy can be used to restore cell function in monogenic disorders or to endow cells with new capabilities, such as improved killing of cancer cells, expression of suicide genes for controlled elimination of cell populations, or protection against chemotherapy or viral infection. While gene therapies were originally most often used to treat monogenic diseases and to improve hematopoietic stem cell transplantation outcome, the advent of genetically modified immune cell therapies, such as chimeric antigen receptor modified T cells, has contributed to the increased numbers of patients treated with gene and cell therapies. The advancement of gene therapy with integrating retroviral vectors continues to depend upon world-wide efforts. As the topic of this special issue is "Spotlight on Germany," the goal of this review is to provide an overview of contributions to this field made by German clinical and research institutions. Research groups in Germany made, and continue to make, important contributions to the development of gene therapy, including design of vectors and transduction protocols for improved cell modification, methods to assess gene therapy vector efficacy and safety (e.g., clonal imbalance, insertion sites), as well as in the design and conduction of clinical gene therapy trials.
Collapse
Affiliation(s)
- Michael A Morgan
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Melanie Galla
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Manuel Grez
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Germany
| | - Boris Fehse
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany.
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4
|
Brendel C, Rio P, Verhoeyen E. Humanized mice are precious tools for evaluation of hematopoietic gene therapies and preclinical modeling to move towards a clinical trial. Biochem Pharmacol 2019; 174:113711. [PMID: 31726047 DOI: 10.1016/j.bcp.2019.113711] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 11/07/2019] [Indexed: 12/11/2022]
Abstract
Over the last decade, incrementally improved xenograft mouse models, which support the engraftment and development of a human hemato-lymphoid system, have been developed and represent an important fundamental and preclinical research tool. Immunodeficient mice can be transplanted with human hematopoietic stem cells (HSCs) and this process is accompanied by HSC homing to the murine bone marrow. This is followed by stem cell expansion, multilineage hematopoiesis, long-term engraftment, and functional human antibody and cellular immune responses. The most significant contributions made by these humanized mice are the identification of normal and leukemic hematopoietic stem cells, the characterization of the human hematopoietic hierarchy, screening of anti-cancer therapies and their use as preclinical models for gene therapy applications. This review article focuses on several gene therapy applications that have benefited from evaluation in humanized mice such as chimeric antigen receptor (CAR) T cell therapies for cancer, anti-viral therapies and gene therapies for multiple monogenetic diseases. Humanized mouse models have been and still are of great value for the gene therapy field since they provide a more reliable understanding of sometimes complicated therapeutic approaches such as recently developed therapeutic gene editing strategies, which seek to correct a gene at its endogenous genomic locus. Additionally, humanized mouse models, which are of great importance with regard to testing new vector technologies in vivo for assessing safety and efficacy prior toclinical trials, help to expedite the critical translation from basic findings to clinical applications. In this review, innovative gene therapies and preclinical studies to evaluate T- and B-cell and HSC-based therapies in humanized mice are discussed and illustrated by multiple examples.
Collapse
Affiliation(s)
- Christian Brendel
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Paula Rio
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | - Els Verhoeyen
- CIRI, Université de Lyon, INSERM U1111, ENS de Lyon, Université Lyon1, CNRS, UMR 5308, 69007 Lyon, France; Université Côte d'Azur, INSERM, C3M, 06204 Nice, France.
| |
Collapse
|
5
|
Abstract
In the wake of a breakthrough in biotechnology providing realistic application of recombinant expressed proteins as drugs in the 1990s, gene therapy emerged as the potential approach for providing medicines of the future [...].
Collapse
|
6
|
RNA Viruses as Tools in Gene Therapy and Vaccine Development. Genes (Basel) 2019; 10:genes10030189. [PMID: 30832256 PMCID: PMC6471356 DOI: 10.3390/genes10030189] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 02/19/2019] [Accepted: 02/21/2019] [Indexed: 12/11/2022] Open
Abstract
RNA viruses have been subjected to substantial engineering efforts to support gene therapy applications and vaccine development. Typically, retroviruses, lentiviruses, alphaviruses, flaviviruses rhabdoviruses, measles viruses, Newcastle disease viruses, and picornaviruses have been employed as expression vectors for treatment of various diseases including different types of cancers, hemophilia, and infectious diseases. Moreover, vaccination with viral vectors has evaluated immunogenicity against infectious agents and protection against challenges with pathogenic organisms. Several preclinical studies in animal models have confirmed both immune responses and protection against lethal challenges. Similarly, administration of RNA viral vectors in animals implanted with tumor xenografts resulted in tumor regression and prolonged survival, and in some cases complete tumor clearance. Based on preclinical results, clinical trials have been conducted to establish the safety of RNA virus delivery. Moreover, stem cell-based lentiviral therapy provided life-long production of factor VIII potentially generating a cure for hemophilia A. Several clinical trials on cancer patients have generated anti-tumor activity, prolonged survival, and even progression-free survival.
Collapse
|
7
|
Lundstrom K. Viral Vectors in Gene Therapy. Diseases 2018; 6:diseases6020042. [PMID: 29883422 PMCID: PMC6023384 DOI: 10.3390/diseases6020042] [Citation(s) in RCA: 303] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/15/2018] [Accepted: 05/16/2018] [Indexed: 01/02/2023] Open
Abstract
Applications of viral vectors have found an encouraging new beginning in gene therapy in recent years. Significant improvements in vector engineering, delivery, and safety have placed viral vector-based therapy at the forefront of modern medicine. Viral vectors have been employed for the treatment of various diseases such as metabolic, cardiovascular, muscular, hematologic, ophthalmologic, and infectious diseases and different types of cancer. Recent development in the area of immunotherapy has provided both preventive and therapeutic approaches. Furthermore, gene silencing generating a reversible effect has become an interesting alternative, and is well-suited for delivery by viral vectors. A number of preclinical studies have demonstrated therapeutic and prophylactic efficacy in animal models and furthermore in clinical trials. Several viral vector-based drugs have also been globally approved.
Collapse
|
8
|
Peking P, Koller U, Duarte B, Murillas R, Wolf S, Maetzig T, Rothe M, Kocher T, García M, Brachtl G, Schambach A, Larcher F, Reichelt J, Bauer JW, Murauer EM. An RNA-targeted therapy for dystrophic epidermolysis bullosa. Nucleic Acids Res 2017; 45:10259-10269. [PMID: 28973459 PMCID: PMC5737646 DOI: 10.1093/nar/gkx669] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 07/20/2017] [Indexed: 11/14/2022] Open
Abstract
Functional impairment or complete loss of type VII collagen, caused by mutations within COL7A1, lead to the severe recessive form of the skin blistering disease dystrophic epidermolysis bullosa (RDEB). Here, we successfully demonstrate RNA trans-splicing as an auspicious repair option for mutations located in a wide range of exons by fully converting an RDEB phenotype in an ex vivo pre-clinical mouse model based on xenotransplantation. Via a self-inactivating (SIN) lentiviral vector a 3' RNA trans-splicing molecule, capable of replacing COL7A1 exons 65-118, was delivered into type VII collagen deficient patient keratinocytes, carrying a homozygous mutation in exon 80 (c.6527insC). Following vector integration, protein analysis of an isolated corrected single cell clone showed secretion of the corrected type VII collagen at similar levels compared to normal keratinocytes. To confirm full phenotypic and long-term correction in vivo, patches of skin equivalents expanded from the corrected cell clone were grafted onto immunodeficient mice. Immunolabelling of 12 weeks old skin specimens showed strong expression of human type VII collagen restricted to the basement membrane zone. We demonstrate that the RNA trans-splicing technology combined with a SIN lentiviral vector is suitable for an ex vivo molecular therapy approach and thus adaptable for clinical application.
Collapse
Affiliation(s)
- Patricia Peking
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology, University Hospital of the Paracelsus Medical University Salzburg, Austria
| | - Ulrich Koller
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology, University Hospital of the Paracelsus Medical University Salzburg, Austria
| | - Blanca Duarte
- Epithelial Biomedicine Division, CIEMAT-CIBERER, Department of Bioengineering, UC3M, Madrid, Spain
| | - Rodolfo Murillas
- Epithelial Biomedicine Division, CIEMAT-CIBERER, Department of Bioengineering, UC3M, Madrid, Spain
| | - Susanne Wolf
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Tobias Maetzig
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Michael Rothe
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Thomas Kocher
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology, University Hospital of the Paracelsus Medical University Salzburg, Austria
| | - Marta García
- Epithelial Biomedicine Division, CIEMAT-CIBERER, Department of Bioengineering, UC3M, Madrid, Spain
| | - Gabriele Brachtl
- Institute for Experimental and Clinical Cell Therapy, Core Facility for Flow Cytometry, SCI-TRECS, Paracelsus Medical University, Salzburg, Austria
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Fernando Larcher
- Epithelial Biomedicine Division, CIEMAT-CIBERER, Department of Bioengineering, UC3M, Madrid, Spain.,Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Madrid, Spain
| | - Julia Reichelt
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology, University Hospital of the Paracelsus Medical University Salzburg, Austria
| | - Johann W Bauer
- Department of Dermatology, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Eva M Murauer
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology, University Hospital of the Paracelsus Medical University Salzburg, Austria
| |
Collapse
|
9
|
Kohlscheen S, Bonig H, Modlich U. Promises and Challenges in Hematopoietic Stem Cell Gene Therapy. Hum Gene Ther 2017; 28:782-799. [DOI: 10.1089/hum.2017.141] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Affiliation(s)
- Saskia Kohlscheen
- Research Group for Gene Modification in Stem Cells, Center for Cell and Gene Therapy Frankfurt, Paul-Ehrlich-Institute, Langen, Germany
| | - Halvard Bonig
- Institute for Transfusion Medicine and Immunohematology, Goethe University, Frankfurt, Germany
- German Red Cross Blood Service Baden-Württemberg-Hessen, Institute Frankfurt, Germany
- Department of Medicine/Division of Hematology, University of Washington, Seattle, Washington
| | - Ute Modlich
- Research Group for Gene Modification in Stem Cells, Center for Cell and Gene Therapy Frankfurt, Paul-Ehrlich-Institute, Langen, Germany
| |
Collapse
|
10
|
Peacock ME, Arce RM, Cutler CW. Periodontal and other oral manifestations of immunodeficiency diseases. Oral Dis 2017; 23:866-888. [PMID: 27630012 PMCID: PMC5352551 DOI: 10.1111/odi.12584] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 08/31/2016] [Accepted: 09/08/2016] [Indexed: 12/14/2022]
Abstract
The list of immunodeficiency diseases grows each year as novel disorders are discovered, classified, and sometimes reclassified due to our ever-increasing knowledge of immune system function. Although the number of patients with secondary immunodeficiencies (SIDs) greatly exceeds those with primary immunodeficiencies (PIDs), the prevalence of both appears to be on the rise probably because of scientific breakthroughs that facilitate earlier and more accurate diagnosis. Primary immunodeficiencies in adults are not as rare as once thought. Globally, the main causes of secondary immunodeficiency are HIV infection and nutritional insufficiencies. Persons with acquired immune disorders such as AIDS caused by the human immunodeficiency virus (HIV) are now living long and fulfilling lives as a result of highly active antiretroviral therapy (HAART). Irrespective of whether the patient's immune-deficient state is a consequence of a genetic defect or is secondary in nature, dental and medical practitioners must be aware of the constant potential for infections and/or expressions of autoimmunity in these individuals. The purpose of this review was to study the most common conditions resulting from primary and secondary immunodeficiency states, how they are classified, and the detrimental manifestations of these disorders on the periodontal and oral tissues.
Collapse
Affiliation(s)
- Mark E Peacock
- Associate Professor, Departments of Periodontics, Oral Biology
| | - Roger M. Arce
- Assistant Professor, Departments of Periodontics, Oral Biology
| | - Christopher W Cutler
- Professor, Departments of Periodontics, Oral Biology; Chair, Department of Periodontics, Associate Dean for Research, The Dental College of Georgia at Augusta University
| |
Collapse
|
11
|
Desoubeaux G, Cray C. Rodent Models of Invasive Aspergillosis due to Aspergillus fumigatus: Still a Long Path toward Standardization. Front Microbiol 2017; 8:841. [PMID: 28559881 PMCID: PMC5432554 DOI: 10.3389/fmicb.2017.00841] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 04/24/2017] [Indexed: 01/09/2023] Open
Abstract
Invasive aspergillosis has been studied in laboratory by the means of plethora of distinct animal models. They were developed to address pathophysiology, therapy, diagnosis, or miscellaneous other concerns associated. However, there are great discrepancies regarding all the experimental variables of animal models, and a thorough focus on them is needed. This systematic review completed a comprehensive bibliographic analysis specifically-based on the technical features of rodent models infected with Aspergillus fumigatus. Out the 800 articles reviewed, it was shown that mice remained the preferred model (85.8% of the referenced reports), above rats (10.8%), and guinea pigs (3.8%). Three quarters of the models involved immunocompromised status, mainly by steroids (44.4%) and/or alkylating drugs (42.9%), but only 27.7% were reported to receive antibiotic prophylaxis to prevent from bacterial infection. Injection of spores (30.0%) and inhalation/deposition into respiratory airways (66.9%) were the most used routes for experimental inoculation. Overall, more than 230 distinct A. fumigatus strains were used in models. Of all the published studies, 18.4% did not mention usage of any diagnostic tool, like histopathology or mycological culture, to control correct implementation of the disease and to measure outcome. In light of these findings, a consensus discussion should be engaged to establish a minimum standardization, although this may not be consistently suitable for addressing all the specific aspects of invasive aspergillosis.
Collapse
Affiliation(s)
- Guillaume Desoubeaux
- Division of Comparative Pathology, Department of Pathology and Laboratory Medicine, Miller School of Medicine, University of MiamiMiami, FL, USA.,Service de Parasitologie-Mycologie-Médecine tropicale, Centre Hospitalier Universitaire de ToursTours, France.,Centre d'Etude des Pathologies Respiratoires (CEPR) Institut National de la Santé et de la Recherche Médicale U1100/Équipe 3, Université François-RabelaisTours, France
| | - Carolyn Cray
- Division of Comparative Pathology, Department of Pathology and Laboratory Medicine, Miller School of Medicine, University of MiamiMiami, FL, USA
| |
Collapse
|
12
|
El Ashkar S, Van Looveren D, Schenk F, Vranckx LS, Demeulemeester J, De Rijck J, Debyser Z, Modlich U, Gijsbers R. Engineering Next-Generation BET-Independent MLV Vectors for Safer Gene Therapy. MOLECULAR THERAPY-NUCLEIC ACIDS 2017. [PMID: 28624199 PMCID: PMC5415309 DOI: 10.1016/j.omtn.2017.04.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Retroviral vectors have shown their curative potential in clinical trials correcting monogenetic disorders. However, therapeutic benefits were compromised due to vector-induced dysregulation of cellular genes and leukemia development in a subset of patients. Bromodomain and extraterminal domain (BET) proteins act as cellular cofactors that tether the murine leukemia virus (MLV) pre-integration complex to host chromatin via interaction with the MLV integrase (IN) and thereby define the typical gammaretroviral integration distribution. We engineered next-generation BET-independent (Bin) MLV vectors to retarget their integration to regions where they are less likely to dysregulate nearby genes. We mutated MLV IN to uncouple BET protein interaction and fused it with chromatin-binding peptides. The addition of the CBX1 chromodomain to MLV INW390A efficiently targeted integration away from gene regulatory elements. The retargeted vector produced at high titers and efficiently transduced CD34+ hematopoietic stem cells, while fewer colonies were detected in a serial colony-forming assay, a surrogate test for genotoxicity. Our findings underscore the potential of the engineered vectors to reduce the risk of insertional mutagenesis without compromising transduction efficiency. Ultimately, combined with other safety features in vector design, next-generation BinMLV vectors can improve the safety of gammaretroviral vectors for gene therapy.
Collapse
Affiliation(s)
- Sara El Ashkar
- Laboratory for Molecular Virology and Drug Discovery, Department of Pharmaceutical and Pharmacological Sciences, 3000 Leuven, KU Leuven, Belgium
| | - Dominique Van Looveren
- Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Franziska Schenk
- RG Gene Modification in Stem Cells, LOEWE Center for Cell and Gene Therapy Frankfurt, Paul-Ehrlich-Institute, 63225 Langen, Germany
| | - Lenard S Vranckx
- Laboratory for Molecular Virology and Drug Discovery, Department of Pharmaceutical and Pharmacological Sciences, 3000 Leuven, KU Leuven, Belgium
| | - Jonas Demeulemeester
- Laboratory for Molecular Virology and Drug Discovery, Department of Pharmaceutical and Pharmacological Sciences, 3000 Leuven, KU Leuven, Belgium
| | - Jan De Rijck
- Laboratory for Molecular Virology and Drug Discovery, Department of Pharmaceutical and Pharmacological Sciences, 3000 Leuven, KU Leuven, Belgium
| | - Zeger Debyser
- Laboratory for Molecular Virology and Drug Discovery, Department of Pharmaceutical and Pharmacological Sciences, 3000 Leuven, KU Leuven, Belgium
| | - Ute Modlich
- RG Gene Modification in Stem Cells, LOEWE Center for Cell and Gene Therapy Frankfurt, Paul-Ehrlich-Institute, 63225 Langen, Germany
| | - Rik Gijsbers
- Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium; Leuven Viral Vector Core, KU Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
13
|
Cavazzana M, Six E, Lagresle-Peyrou C, André-Schmutz I, Hacein-Bey-Abina S. Gene Therapy for X-Linked Severe Combined Immunodeficiency: Where Do We Stand? Hum Gene Ther 2016; 27:108-16. [PMID: 26790362 DOI: 10.1089/hum.2015.137] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
More than 20 years ago, X-linked severe combined immunodeficiency (SCID-X1) appeared to be the best condition to test the feasibility of hematopoietic stem cell gene therapy. The seminal SCID-X1 clinical studies, based on first-generation gammaretroviral vectors, demonstrated good long-term immune reconstitution in most treated patients despite the occurrence of vector-related leukemia in a few of them. This gene therapy has successfully enabled correction of the T cell defect. Natural killer and B cell defects were only partially restored, most likely due to the absence of a conditioning regimen. The success of these pioneering trials paved the way for the extension of gene-based treatment to many other diseases of the hematopoietic system, but the unfortunate serious adverse events led to extensive investigations to define the retrovirus integration profiles. This review puts into perspective the clinical experience of gene therapy for SCID-X1, with the development and implementation of new generations of safer vectors such as self-inactivating gammaretroviral or lentiviral vectors as well as major advances in integrome knowledge.
Collapse
Affiliation(s)
- Marina Cavazzana
- 1 Biotherapy Department, Necker Children's Hospital , Assistance Publique-Hôpitaux de Paris, Paris.,2 Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest , Assistance Publique-Hôpitaux de Paris, INSERM, Paris.,3 Paris Descartes-Sorbonne Paris Cité University, Imagine Institute , Paris.,4 INSERM UMR 1163, Laboratory of Human Lymphohematopoiesis , Paris
| | - Emmanuelle Six
- 2 Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest , Assistance Publique-Hôpitaux de Paris, INSERM, Paris.,3 Paris Descartes-Sorbonne Paris Cité University, Imagine Institute , Paris.,4 INSERM UMR 1163, Laboratory of Human Lymphohematopoiesis , Paris
| | - Chantal Lagresle-Peyrou
- 2 Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest , Assistance Publique-Hôpitaux de Paris, INSERM, Paris.,3 Paris Descartes-Sorbonne Paris Cité University, Imagine Institute , Paris.,4 INSERM UMR 1163, Laboratory of Human Lymphohematopoiesis , Paris
| | - Isabelle André-Schmutz
- 2 Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest , Assistance Publique-Hôpitaux de Paris, INSERM, Paris.,3 Paris Descartes-Sorbonne Paris Cité University, Imagine Institute , Paris.,4 INSERM UMR 1163, Laboratory of Human Lymphohematopoiesis , Paris
| | - Salima Hacein-Bey-Abina
- 1 Biotherapy Department, Necker Children's Hospital , Assistance Publique-Hôpitaux de Paris, Paris.,2 Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest , Assistance Publique-Hôpitaux de Paris, INSERM, Paris.,5 UTCBS CNRS 8258-INSERM U1022, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes , Paris.,6 Immunology Laboratory, Groupe Hospitalier Universitaire Paris-Sud , AP-HP, Le-Kremlin-Bicêtre, France
| |
Collapse
|
14
|
Fronza R, Vasciaveo A, Benso A, Schmidt M. A Graph Based Framework to Model Virus Integration Sites. Comput Struct Biotechnol J 2016; 14:69-77. [PMID: 27257470 PMCID: PMC4874582 DOI: 10.1016/j.csbj.2015.10.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 10/20/2015] [Accepted: 10/23/2015] [Indexed: 12/03/2022] Open
Abstract
With next generation sequencing thousands of virus and viral vector integration genome targets are now under investigation to uncover specific integration preferences and to define clusters of integration, termed common integration sites (CIS), that may allow to assess gene therapy safety or to detect disease related genomic features such as oncogenes. Here, we addressed the challenge to: 1) define the notion of CIS on graph models, 2) demonstrate that the structure of CIS enters in the category of scale-free networks and 3) show that our network approach analyzes CIS dynamically in an integrated systems biology framework using the Retroviral Transposon Tagged Cancer Gene Database (RTCGD) as a testing dataset.
Collapse
Affiliation(s)
- Raffaele Fronza
- Department of Translational Oncology, National Center for Tumor Diseases and German Cancer Research Center, Im Neuenheimer Feld 581, 69120 Heidelberg, Germany
| | - Alessandro Vasciaveo
- Department of Translational Oncology, National Center for Tumor Diseases and German Cancer Research Center, Im Neuenheimer Feld 581, 69120 Heidelberg, Germany; Department of Control and Computer Engineering, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Alfredo Benso
- Department of Control and Computer Engineering, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Manfred Schmidt
- Department of Translational Oncology, National Center for Tumor Diseases and German Cancer Research Center, Im Neuenheimer Feld 581, 69120 Heidelberg, Germany
| |
Collapse
|
15
|
LIU ZHAOLIANG, YOU CAILIAN, WANG BIAO, LIN JIANHONG, HU XUEFENG, SHAN XIUYING, WANG MEISHUI, ZHENG HOUBING, ZHANG YANDING. Construction of Ang2-siRNA chitosan magnetic nanoparticles and the effect on Ang2 gene expression in human malignant melanoma cells. Oncol Lett 2016; 11:3992-3998. [PMID: 27313729 PMCID: PMC4888209 DOI: 10.3892/ol.2016.4539] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 03/01/2016] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to construct angiopoietin-2 (Ang2)-small interfering (si)RNA chitosan magnetic nanoparticles and to observe the interference effects of the nanoparticles on the expression of the Ang2 gene in human malignant melanoma cells. Ang2-siRNA chitosan magnetic nanoparticles were constructed and transfected into human malignant melanoma cells in vitro. Red fluorescent protein expression was observed, and the transfection efficiency was analyzed. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was used to assess the inhibition efficiency of Ang2 gene expression. Ang2-siRNA chitosan magnetic nanoparticles were successfully constructed, and at a mass ratio of plasmid to magnetic chitosan nanoparticles of 1:100, the transfection efficiency into human malignant melanoma cells was the highest of the ratios assessed, reaching 61.17%. RT-qPCR analysis showed that the magnetic chitosan nanoparticles effectively inhibited Ang2 gene expression in cells, and the inhibition efficiency reached 59.56% (P<0.05). Ang2-siRNA chitosan magnetic nanoparticles were successfully constructed. The in vitro studies showed that the nanoparticles inhibited Ang2 gene expression in human malignant melanoma tumor cells, which laid the foundation and provided experimental evidence for additional future in vivo studies of intervention targeting malignant melanoma tumor growth in nude mice.
Collapse
Affiliation(s)
- ZHAO-LIANG LIU
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - CAI-LIAN YOU
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - BIAO WANG
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - JIAN-HONG LIN
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - XUE-FENG HU
- College of Life Sciences, Fujian Normal University, Fuzhou, Fujian 350108, P.R. China
| | - XIU-YING SHAN
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - MEI-SHUI WANG
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - HOU-BING ZHENG
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - YAN-DING ZHANG
- College of Life Sciences, Fujian Normal University, Fuzhou, Fujian 350108, P.R. China
| |
Collapse
|
16
|
Booth C, Gaspar HB, Thrasher AJ. Treating Immunodeficiency through HSC Gene Therapy. Trends Mol Med 2016; 22:317-327. [PMID: 26993219 DOI: 10.1016/j.molmed.2016.02.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 02/15/2016] [Accepted: 02/16/2016] [Indexed: 11/19/2022]
Abstract
Haematopoietic stem cell (HSC) gene therapy has been successfully employed as a therapeutic option to treat specific inherited immune deficiencies, including severe combined immune deficiencies (SCID) over the past two decades. Initial clinical trials using first-generation gamma-retroviral vectors to transfer corrective DNA demonstrated clinical benefit for patients, but were associated with leukemogenesis in a number of cases. Safer vectors have since been developed, affording comparable efficacy with an improved biosafety profile. These vectors are now in Phase I/II clinical trials for a number of immune disorders with more preclinical studies underway. Targeted gene editing allowing precise DNA correction via platforms such as ZFNs, TALENs and CRISPR/Cas9 may now offer promising strategies to improve the safety and efficacy of gene therapy in the future.
Collapse
Affiliation(s)
- Claire Booth
- Molecular and Cellular Immunology Section, UCL Institute of Child Health, London, UK; Department of Paediatric Immunology, Great Ormond Street Hospital, London, UK
| | - H Bobby Gaspar
- Molecular and Cellular Immunology Section, UCL Institute of Child Health, London, UK; Department of Paediatric Immunology, Great Ormond Street Hospital, London, UK
| | - Adrian J Thrasher
- Molecular and Cellular Immunology Section, UCL Institute of Child Health, London, UK; Department of Paediatric Immunology, Great Ormond Street Hospital, London, UK.
| |
Collapse
|
17
|
Clonal Dominance With Retroviral Vector Insertions Near the ANGPT1 and ANGPT2 Genes in a Human Xenotransplant Mouse Model. MOLECULAR THERAPY-NUCLEIC ACIDS 2014; 3:e200. [PMID: 25291142 PMCID: PMC4217076 DOI: 10.1038/mtna.2014.51] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 08/10/2014] [Indexed: 12/22/2022]
Abstract
Insertional leukemogenesis represents the major risk factor of hematopoietic stem cell (HSC) based gene therapy utilizing integrating viral vectors. To develop a pre-clinical model for the evaluation of vector-related genotoxicity directly in the relevant human target cells, cord blood CD34+ HSCs were transplanted into immunodeficient NOD.SCID.IL2rg−/− (NSG) mice after transduction with an LTR-driven gammaretroviral vector (GV). Furthermore, we specifically investigated the effect of prolonged in vitro culture in the presence of cytokines recently described to promote HSC expansion or maintenance. Clonality of human hematopoiesis in NSG mice was assessed by high throughput insertion site analyses and validated by insertion site-specific PCR depicting a GV typical integration profile with insertion sites resembling to 25% those of clinical studies. No overrepresentation of integrations in the vicinity of cancer-related genes was observed, however, several dominant clones were identified including two clones harboring integrations in the ANGPT1 and near the ANGPT2 genes associated with deregulated ANGPT1- and ANGPT2-mRNA levels. While these data underscore the potential value of the NSG model, our studies also identified short-comings such as overall low numbers of engrafted HSCs, limited in vivo observation time, and the challenges of in-depth insertion site analyses by low contribution of gene modified hematopoiesis.
Collapse
|
18
|
Dighe N, Khoury M, Mattar C, Chong M, Choolani M, Chen J, Antoniou MN, Chan JKY. Long-term reproducible expression in human fetal liver hematopoietic stem cells with a UCOE-based lentiviral vector. PLoS One 2014; 9:e104805. [PMID: 25118036 PMCID: PMC4130605 DOI: 10.1371/journal.pone.0104805] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 07/14/2014] [Indexed: 12/31/2022] Open
Abstract
Hematopoietic Stem Cell (HSC) targeted gene transfer is an attractive treatment option for a number of hematopoietic disorders caused by single gene defects. However, extensive methylation of promoter sequences results in silencing of therapeutic gene expression. The choice of an appropriate promoter is therefore crucial for reproducible, stable and long-term transgene expression in clinical gene therapy. Recent studies suggest efficient and stable expression of transgenes from the ubiquitous chromatin opening element (UCOE) derived from the human HNRPA2B1-CBX3 locus can be achieved in murine HSC. Here, we compared the use of HNRPA2B1-CBX3 UCOE (A2UCOE)-mediated transgene regulation to two other frequently used promoters namely EF1α and PGK in human fetal liver-derived HSC (hflHSC). Efficient transduction of hflHSC with a lentiviral vector containing an HNRPA2B1-CBX3 UCOE-eGFP (A2UCOE-eGFP) cassette was achieved at higher levels than that obtained with umbilical cord blood derived HSC (3.1x; p<0.001). While hflHSC were readily transduced with all three test vectors (A2UCOE-eGFP, PGK-eGFP and EF1α-eGFP), only the A2-UCOE construct demonstrated sustained transgene expression in vitro over 24 days (p<0.001). In contrast, within 10 days in culture a rapid decline in transgene expression in both PGK-eGFP and EF1α-eGFP transduced hflHSC was seen. Subsequently, injection of transduced cells into immunodeficient mice (NOD/SCID/Il2rg-/-) demonstrated sustained eGFP expression for the A2UCOE-eGFP group up to 10 months post transplantation whereas PGK-eGFP and EF1α-eGFP transduced hflHSC showed a 5.1 and 22.2 fold reduction respectively over the same time period. We conclude that the A2UCOE allows a more efficient and stable expression in hflHSC to be achieved than either the PGK or EF1α promoters and at lower vector copy number per cell.
Collapse
Affiliation(s)
- Niraja Dighe
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Maroun Khoury
- Interdisciplinary Research Group in Infectious Diseases, Singapore-Massachusetts Institute of Technology Alliance for Research and Technology, Singapore, Singapore
| | - Citra Mattar
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mark Chong
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Bioengineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | - Mahesh Choolani
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jianzhu Chen
- Interdisciplinary Research Group in Infectious Diseases, Singapore-Massachusetts Institute of Technology Alliance for Research and Technology, Singapore, Singapore
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Michael N. Antoniou
- Department of Medical and Molecular Genetics, King's College London School of Medicine, Guys Hospital, London, United Kingdom
| | - Jerry K. Y. Chan
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore
- Cancer and Stem Cell Program, Duke-NUS Graduate Medical School, Singapore, Singapore
- * E-mail:
| |
Collapse
|
19
|
Warris A, Henriet SSV. Invasive Fungal Infections in the Child with Chronic Granulomatous Disease. CURRENT FUNGAL INFECTION REPORTS 2014. [DOI: 10.1007/s12281-013-0168-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
20
|
Gunzer M. Traps and hyper inflammation - new ways that neutrophils promote or hinder survival. Br J Haematol 2013; 164:189-99. [PMID: 24138538 DOI: 10.1111/bjh.12608] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 09/11/2013] [Indexed: 01/13/2023]
Abstract
For a long time neutrophil granulocytes were considered simply as terminally differentiated cells with a limited life span and pathogen killing by phagocytosis and chemical toxicity being the sole mode of action. However, work during the last 10 years has started to change this view fundamentally. Modern understanding is that neutrophils have an enormous complexity of functions. This review discusses very recent findings on how neutrophils can control the spread of pathogens and mediate their killing by mechanisms such as formation of DNA nets, how they influence tumour growth and adaptive immune responses and how they manoeuvre inside the diverse compartments of the body. It will also describe how the normally protective functions of neutrophils can have deleterious consequences if they occur in an uncontrolled fashion. These exciting novel findings are likely to completely and permanently change our view of this central leucocyte population.
Collapse
Affiliation(s)
- Matthias Gunzer
- Institute for Experimental Immunology and Imaging, University Duisburg-Essen, University Hospital, Essen, Germany
| |
Collapse
|