251
|
Preclinical Development of Autologous Hematopoietic Stem Cell-Based Gene Therapy for Immune Deficiencies: A Journey from Mouse Cage to Bed Side. Pharmaceutics 2020; 12:pharmaceutics12060549. [PMID: 32545727 PMCID: PMC7357087 DOI: 10.3390/pharmaceutics12060549] [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] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/05/2020] [Accepted: 06/09/2020] [Indexed: 02/08/2023] Open
Abstract
Recent clinical trials using patient’s own corrected hematopoietic stem cells (HSCs), such as for primary immunodeficiencies (Adenosine deaminase (ADA) deficiency, X-linked Severe Combined Immunodeficiency (SCID), X-linked chronic granulomatous disease (CGD), Wiskott–Aldrich Syndrome (WAS)), have yielded promising results in the clinic; endorsing gene therapy to become standard therapy for a number of diseases. However, the journey to achieve such a successful therapy is not easy, and several challenges have to be overcome. In this review, we will address several different challenges in the development of gene therapy for immune deficiencies using our own experience with Recombinase-activating gene 1 (RAG1) SCID as an example. We will discuss product development (targeting of the therapeutic cells and choice of a suitable vector and delivery method), the proof-of-concept (in vitro and in vivo efficacy, toxicology, and safety), and the final release steps to the clinic (scaling up, good manufacturing practice (GMP) procedures/protocols and regulatory hurdles).
Collapse
|
252
|
Afzal S, Fronza R, Schmidt M. VSeq-Toolkit: Comprehensive Computational Analysis of Viral Vectors in Gene Therapy. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 17:752-757. [PMID: 32346552 PMCID: PMC7177155 DOI: 10.1016/j.omtm.2020.03.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 03/25/2020] [Indexed: 11/17/2022]
Abstract
Viral vector characterization and analysis are important components for the development of safe gene therapeutic products, elucidating the potential genotoxic and immunogenic effects of vectors and establishing their safety profiles. Here, we present VSeq-Toolkit, which offers varying analysis modes for viral gene therapy data. The first mode determines the undesirable known contaminants and their frequency in viral preparations or other sequencing data. The second mode is designed for the analysis of intra-vector fusion breakpoints and the third mode for unraveling the viral-host fusion events distribution. Analysis modes of our toolkit can be executed independently or together and allow the analysis of multiple viral vectors concurrently. It has been designed and evaluated for the analysis of short read high-throughput sequencing data, including whole-genome or targeted sequencing. VSeq-Toolkit is developed in Perl and Bash programming languages and is available at https://github.com/CompMeth/VSeq-Toolkit.
Collapse
Affiliation(s)
- Saira Afzal
- Department of Translational Oncology, German Cancer Research Center (DKFZ), National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Corresponding author: Saira Afzal, Department of Translational Oncology (G100), German Cancer Research Center (DKFZ), National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 581, 69120 Heidelberg, Germany.
| | | | - Manfred Schmidt
- Department of Translational Oncology, German Cancer Research Center (DKFZ), National Center for Tumor Diseases (NCT), Heidelberg, Germany
- GeneWerk GmbH, Heidelberg, Germany
| |
Collapse
|
253
|
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: 40] [Impact Index Per Article: 8.0] [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
|
254
|
Humbert O, Samuelson C, Kiem HP. CRISPR/Cas9 for the treatment of haematological diseases: a journey from bacteria to the bedside. Br J Haematol 2020; 192:33-49. [PMID: 32506752 DOI: 10.1111/bjh.16807] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/07/2020] [Accepted: 05/09/2020] [Indexed: 12/26/2022]
Abstract
Genome editing therapies represent a significant advancement in next-generation, precision medicine for the management of haematological diseases, and CRISPR/Cas9 has to date been the most successful implementation platform. From discovery in bacteria and archaea over three decades ago, through intensive basic research and pre-clinical development phases involving the modification of therapeutically relevant cell types, CRISPR/Cas9 genome editing is now being investigated in ongoing clinic trials. Despite the widespread enthusiasm brought by this new technology, significant challenges remain before genome editing can be routinely recommended and implemented in the clinic. These include risks of genotoxicity resulting from off-target DNA cleavage or chromosomal rearrangement, and suboptimal efficacy of homology-directed repair editing strategies, which thus limit therapeutic options. Practical hurdles such as high costs and inaccessibility to patients outside specialised centres must also be addressed. Future improvements in this rapidly developing field should circumvent current limitations with novel editing platforms and with the simplification of clinical protocols using in vivo delivery of editing reagents.
Collapse
Affiliation(s)
| | | | - Hans-Peter Kiem
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
255
|
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
|
256
|
Milton-Harris L, Jeeves M, Walker SA, Ward SE, Mancini EJ. Small molecule inhibits T-cell acute lymphoblastic leukaemia oncogenic interaction through conformational modulation of LMO2. Oncotarget 2020; 11:1737-1748. [PMID: 32477463 PMCID: PMC7233811 DOI: 10.18632/oncotarget.27580] [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] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 04/03/2020] [Indexed: 01/05/2023] Open
Abstract
Ectopic expression in T-cell precursors of LIM only protein 2 (LMO2), a key factor in hematopoietic development, has been linked to the onset of T-cell acute lymphoblastic leukaemia (T-ALL). In the T-ALL context, LMO2 drives oncogenic progression through binding to erythroid-specific transcription factor SCL/TAL1 and sequestration of E-protein transcription factors, normally required for T-cell differentiation. A key requirement for the formation of this oncogenic protein-protein interaction (PPI) is the conformational flexibility of LMO2. Here we identify a small molecule inhibitor of the SCL-LMO2 PPI, which hinders the interaction in vitro through direct binding to LMO2. Biophysical analysis demonstrates that this inhibitor acts through a mechanism of conformational modulation of LMO2. Importantly, this work has led to the identification of a small molecule inhibitor of the SCL-LMO2 PPI, which can provide a starting point for the development of new agents for the treatment of T-ALL. These results suggest that similar approaches, based on the modulation of protein conformation by small molecules, might be used for therapeutic targeting of other oncogenic PPIs.
Collapse
Affiliation(s)
- Leanne Milton-Harris
- School of Life Sciences, Biochemistry Department, University of Sussex, Falmer, Brighton, BN1 9QG, United Kingdom
| | - Mark Jeeves
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom
| | - Sarah A Walker
- Sussex Drug Discovery Centre, University of Sussex, Brighton, BN1 9QJ, United Kingdom
| | - Simon E Ward
- Medicines Discovery Institute, Cardiff University, Park Place, Cardiff, CF10 3AT, United Kingdom
| | - Erika J Mancini
- School of Life Sciences, Biochemistry Department, University of Sussex, Falmer, Brighton, BN1 9QG, United Kingdom
| |
Collapse
|
257
|
Stok M, de Boer H, Huston MW, Jacobs EH, Roovers O, Visser TP, Jahr H, Duncker DJ, van Deel ED, Reuser AJJ, van Til NP, Wagemaker G. Lentiviral Hematopoietic Stem Cell Gene Therapy Corrects Murine Pompe Disease. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 17:1014-1025. [PMID: 32462050 PMCID: PMC7240064 DOI: 10.1016/j.omtm.2020.04.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 04/27/2020] [Indexed: 01/07/2023]
Abstract
Pompe disease is an autosomal recessive lysosomal storage disorder characterized by progressive muscle weakness. The disease is caused by mutations in the acid α-glucosidase (GAA) gene. Despite the currently available enzyme replacement therapy (ERT), roughly half of the infants with Pompe disease die before the age of 3 years. Limitations of ERT are immune responses to the recombinant enzyme, incomplete correction of the disease phenotype, lifelong administration, and inability of the enzyme to cross the blood-brain barrier. We previously reported normalization of glycogen in heart tissue and partial correction of the skeletal muscle phenotype by ex vivo hematopoietic stem cell gene therapy. In the present study, using a codon-optimized GAA (GAAco), the enzyme levels resulted in close to normalization of glycogen in heart, muscles, and brain, and in complete normalization of motor function. A large proportion of microglia in the brain was shown to be GAA positive. All astrocytes contained the enzyme, which is in line with mannose-6-phosphate receptor expression and the key role in glycogen storage and glucose metabolism. The lentiviral vector insertion site analysis confirmed no preference for integration near proto-oncogenes. This correction of murine Pompe disease warrants further development toward a cure of the human condition.
Collapse
Affiliation(s)
- Merel Stok
- Department of Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Pediatrics, Erasmus University Medical Center, Rotterdam, the Netherlands.,Center for Lysosomal and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Helen de Boer
- Department of Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands.,Molecular Stem Cell Biology, Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marshall W Huston
- Department of Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Edwin H Jacobs
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Onno Roovers
- Department of Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Trudi P Visser
- Department of Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Holger Jahr
- Department of Orthopaedics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Elza D van Deel
- Division of Experimental Cardiology, Department of Cardiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Arnold J J Reuser
- Center for Lysosomal and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands.,Molecular Stem Cell Biology, Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Niek P van Til
- Department of Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Gerard Wagemaker
- Department of Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
258
|
Bruter AV, Kalashnikova MV, Prytyko AP, Belyavsky AV. Maintenance of Plasmid Expression in vivo Depends Primarily on the CpG Contents of the Vector and Transgene. Mol Biol 2020; 54:427-435. [DOI: 10.1134/s0026893320030048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/12/2019] [Accepted: 12/12/2019] [Indexed: 01/04/2025]
|
259
|
De Rosa L, Latella MC, Secone Seconetti A, Cattelani C, Bauer JW, Bondanza S, De Luca M. Toward Combined Cell and Gene Therapy for Genodermatoses. Cold Spring Harb Perspect Biol 2020; 12:a035667. [PMID: 31653644 PMCID: PMC7197428 DOI: 10.1101/cshperspect.a035667] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
To date, more than 200 monogenic, often devastating, skin diseases have been described. Because of unmet medical needs, development of long-lasting and curative therapies has been consistently attempted, with the aim of correcting the underlying molecular defect. In this review, we will specifically address the few combined cell and gene therapy strategies that made it to the clinics. Based on these studies, what can be envisioned for the future is a patient-oriented strategy, built on the specific features of the individual in need. Most likely, a combination of different strategies, approaches, and advanced therapies will be required to reach the finish line at the end of the long and winding road hampering the achievement of definitive treatments for genodermatoses.
Collapse
Affiliation(s)
- Laura De Rosa
- Holostem Terapie Avanzate S.r.l., Center for Regenerative Medicine "Stefano Ferrari," 41125 Modena, Italy
| | - Maria Carmela Latella
- Holostem Terapie Avanzate S.r.l., Center for Regenerative Medicine "Stefano Ferrari," 41125 Modena, Italy
| | - Alessia Secone Seconetti
- Holostem Terapie Avanzate S.r.l., Center for Regenerative Medicine "Stefano Ferrari," 41125 Modena, Italy
| | - Cecilia Cattelani
- Center for Regenerative Medicine "Stefano Ferrari," Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Johann W Bauer
- EB House Austria and Department of Dermatology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Sergio Bondanza
- Holostem Terapie Avanzate S.r.l., Center for Regenerative Medicine "Stefano Ferrari," 41125 Modena, Italy
| | - Michele De Luca
- Center for Regenerative Medicine "Stefano Ferrari," Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| |
Collapse
|
260
|
Santeramo I, Bagnati M, Harvey EJ, Hassan E, Surmacz-Cordle B, Marshall D, Di Cerbo V. Vector Copy Distribution at a Single-Cell Level Enhances Analytical Characterization of Gene-Modified Cell Therapies. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 17:944-956. [PMID: 32420408 PMCID: PMC7217927 DOI: 10.1016/j.omtm.2020.04.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 04/22/2020] [Indexed: 12/28/2022]
Abstract
The ability to deliver transgenes into the human genome using viral vectors is a major enabler of the gene-modified cell therapy field. However, the control of viral transduction is difficult and can lead to product heterogeneity, impacting efficacy and safety, as well as increasing the risk of batch failure during manufacturing. To address this, we generated a novel analytical method to measure vector copy distribution at the single-cell level in a gene-modified, lentiviral-based immunotherapy model. As the limited amount of genomic DNA in a single cell hinders reliable quantification, we implemented a preamplification strategy on selected lentiviral and human gene targets in isolated live single cells, followed by quantification of amplified material by droplet digital PCR. Using a bespoke probability framework based on Bayesian statistics, we show that we can estimate vector copy number (VCN) integers with maximum likelihood scores. Notably, single-cell data are consistent with population analysis and also provide an overall measurement of transduction efficiency by discriminating transduced (VCN ≥ 1) from nontransduced (VCN = 0) cells. The ability to characterize cell-to-cell variability provides a powerful high-resolution approach for product characterization, which could ultimately allow improved control over product quality and safety.
Collapse
Affiliation(s)
- Ilaria Santeramo
- Cell and Gene Therapy Catapult, 12th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom
| | - Marta Bagnati
- Cell and Gene Therapy Catapult, 12th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom
| | - Emily Jane Harvey
- Cell and Gene Therapy Catapult, 12th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom
| | - Enas Hassan
- Cell and Gene Therapy Catapult, 12th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom
| | - Beata Surmacz-Cordle
- Cell and Gene Therapy Catapult, 12th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom
| | - Damian Marshall
- Cell and Gene Therapy Catapult, 12th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom
| | - Vincenzo Di Cerbo
- Cell and Gene Therapy Catapult, 12th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom
| |
Collapse
|
261
|
Butterfield JSS, Hege KM, Herzog RW, Kaczmarek R. A Molecular Revolution in the Treatment of Hemophilia. Mol Ther 2020; 28:997-1015. [PMID: 31843450 PMCID: PMC7132613 DOI: 10.1016/j.ymthe.2019.11.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/31/2019] [Accepted: 11/05/2019] [Indexed: 12/15/2022] Open
Abstract
For decades, the monogenetic bleeding disorders hemophilia A and B (coagulation factor VIII and IX deficiency) have been treated with systemic protein replacement therapy. Now, diverse molecular medicines, ranging from antibody to gene to RNA therapy, are transforming treatment. Traditional replacement therapy requires twice to thrice weekly intravenous infusions of factor. While extended half-life products may reduce the frequency of injections, patients continue to face a lifelong burden of the therapy, suboptimal protection from bleeding and joint damage, and potential development of neutralizing anti-drug antibodies (inhibitors) that require less efficacious bypassing agents and further reduce quality of life. Novel non-replacement and gene therapies aim to address these remaining issues. A recently approved factor VIII-mimetic antibody accomplishes hemostatic correction in patients both with and without inhibitors. Antibodies against tissue factor pathway inhibitor (TFPI) and antithrombin-specific small interfering RNA (siRNA) target natural anticoagulant pathways to rebalance hemostasis. Adeno-associated virus (AAV) gene therapy provides lasting clotting factor replacement and can also be used to induce immune tolerance. Multiple gene-editing techniques are under clinical or preclinical investigation. Here, we provide a comprehensive overview of these approaches, explain how they differ from standard therapies, and predict how the hemophilia treatment landscape will be reshaped.
Collapse
Affiliation(s)
| | - Kerry M Hege
- Department of Pediatrics, Indiana University School of Medicine, IUPUI-Wells Center for Pediatric Research, Indianapolis, IN, USA
| | - Roland W Herzog
- Department of Pediatrics, University of Florida, Gainesville, FL, USA; Department of Pediatrics, Indiana University School of Medicine, IUPUI-Wells Center for Pediatric Research, Indianapolis, IN, USA.
| | - Radoslaw Kaczmarek
- Department of Pediatrics, Indiana University School of Medicine, IUPUI-Wells Center for Pediatric Research, Indianapolis, IN, USA; Laboratory of Glycobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, Poland.
| |
Collapse
|
262
|
Abstract
The technological advances in diagnostics and therapy of primary immunodeficiency are progressing at a fast pace. This review examines recent developments in the field of inborn errors of immunity, from their definition to their treatment. We will summarize the challenges posed by the growth of next-generation sequencing in the clinical setting, touch briefly on the expansion of the concept of inborn errors of immunity beyond the classic immune system realm, and finally review current developments in targeted therapies, stem cell transplantation, and gene therapy.
Collapse
Affiliation(s)
- Giorgia Bucciol
- Inborn Errors of Immunity, Department of Immunology, Microbiology and Transplantation, KU Leuven, Herestraat 49, Leuven, 3000, Belgium.,Childhood Immunology, Department of Pediatrics, University Hospitals Leuven, ERN-RITA Core Member, Herestraat 49, Leuven, 3000, Belgium
| | - Isabelle Meyts
- Inborn Errors of Immunity, Department of Immunology, Microbiology and Transplantation, KU Leuven, Herestraat 49, Leuven, 3000, Belgium.,Childhood Immunology, Department of Pediatrics, University Hospitals Leuven, ERN-RITA Core Member, Herestraat 49, Leuven, 3000, Belgium
| |
Collapse
|
263
|
Enhanced Production of Herpes Simplex Virus 1 Amplicon Vectors by Gene Modification and Optimization of Packaging Cell Growth Medium. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 17:491-496. [PMID: 32258212 PMCID: PMC7114837 DOI: 10.1016/j.omtm.2020.03.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 03/10/2020] [Indexed: 01/25/2023]
Abstract
Herpes simplex virus 1 (HSV-1)-derived amplicon vectors are unique in their ability to accommodate large DNA molecules allowing whole genomic loci to be included with all of their regulatory elements. Additional advantages of these amplicons include their minimal toxicity and ability to persist as episomes, with negligible risk of insertional mutagenesis, being particularly well-suited for gene therapy of neurological disorders due to their outstanding ability to deliver genes into neurons and other neural cells. However, extensive gene therapy application has been hindered by difficulties in vector production. This work improved HSV-1 amplicons production by genetic modification of the packaging cell line and optimization of the culture medium. A stably-transfected Vero 2-2 cell line overexpressing the anti-apoptotic Bcl-2 protein was generated, exhibiting an increased resistance to apoptosis, prolonged culture duration, and a significant improvement in viral vector production. Additionally, supplementation of the growth medium with antioxidants, polyamines, amino acids, and reduced glutathione further increased the yield of packaged amplicon vectors. With these modifications, HSV-1 amplicons could be isolated from culture supernatants instead of cell lysates, leading to vector preparations with higher titer and purity and paving the way for generation of stable cell lines that are capable of continuous herpesviral vector production.
Collapse
|
264
|
Kao RL, Truscott LC, Chiou TT, Tsai W, Wu AM, De Oliveira SN. A Cetuximab-Mediated Suicide System in Chimeric Antigen Receptor-Modified Hematopoietic Stem Cells for Cancer Therapy. Hum Gene Ther 2020; 30:413-428. [PMID: 30860401 DOI: 10.1089/hum.2018.180] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Using gene modification of hematopoietic stem cells (HSC) to create persistent generation of multilineage immune effectors to target cancer cells directly is proposed. Gene-modified human HSC have been used to introduce genes to correct, prevent, or treat diseases. Concerns regarding malignant transformation, abnormal hematopoiesis, and autoimmunity exist, making the co-delivery of a suicide gene a necessary safety measure. Truncated epidermal growth factor receptor (EGFRt) was tested as a suicide gene system co-delivered with anti-CD19 chimeric antigen receptor (CAR) to human HSC. Third-generation self-inactivating lentiviral vectors were used to co-deliver an anti-CD19 CAR and EGFRt. In vitro, gene-modified HSC were differentiated into myeloid cells to allow transgene expression. An antibody-dependent cell-mediated cytotoxicity (ADCC) assay was used, incubating target cells with leukocytes and monoclonal antibody cetuximab to determine the percentage of surviving cells. In vivo, gene-modified HSC were engrafted into NSG mice with subsequent treatment with intraperitoneal cetuximab. Persistence of gene-modified cells was assessed by flow cytometry, droplet digital polymerase chain reaction (ddPCR), and positron emission tomography (PET) imaging using 89Zr-Cetuximab. Cytotoxicity was significantly increased (p = 0.01) in target cells expressing EGFRt after incubation with leukocytes and cetuximab 1 μg/mL compared to EGFRt+ cells without cetuximab and non-transduced cells with or without cetuximab, at all effector:target ratios. Mice humanized with gene-modified HSC presented significant ablation of gene-modified cells after treatment (p = 0.002). Remaining gene-modified cells were close to background on flow cytometry and within two logs of decrease of vector copy numbers by ddPCR in mouse tissues. PET imaging confirmed ablation with a decrease of an average of 82.5% after cetuximab treatment. These results give proof of principle for CAR-modified HSC regulated by a suicide gene. Further studies are needed to enable clinical translation. Cetuximab ADCC of EGFRt-modified cells caused effective killing. Different ablation approaches, such as inducible caspase 9 or co-delivery of other inert cell markers, should also be evaluated.
Collapse
Affiliation(s)
- Roy L Kao
- 1 Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Laurel C Truscott
- 1 Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Tzu-Ting Chiou
- 1 Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Wenting Tsai
- 2 Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California
| | - Anna M Wu
- 2 Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California
| | - Satiro N De Oliveira
- 1 Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|
265
|
Young E, Gould D, Hart S. Toward gene therapy in rheumatoid arthritis. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2020. [DOI: 10.1080/23808993.2020.1736942] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Emily Young
- Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - David Gould
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Stephen Hart
- Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
266
|
Kovač A, Miskey C, Menzel M, Grueso E, Gogol-Döring A, Ivics Z. RNA-guided retargeting of S leeping Beauty transposition in human cells. eLife 2020; 9:e53868. [PMID: 32142408 PMCID: PMC7077980 DOI: 10.7554/elife.53868] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 03/05/2020] [Indexed: 12/12/2022] Open
Abstract
An ideal tool for gene therapy would enable efficient gene integration at predetermined sites in the human genome. Here we demonstrate biased genome-wide integration of the Sleeping Beauty (SB) transposon by combining it with components of the CRISPR/Cas9 system. We provide proof-of-concept that it is possible to influence the target site selection of SB by fusing it to a catalytically inactive Cas9 (dCas9) and by providing a single guide RNA (sgRNA) against the human Alu retrotransposon. Enrichment of transposon integrations was dependent on the sgRNA, and occurred in an asymmetric pattern with a bias towards sites in a relatively narrow, 300 bp window downstream of the sgRNA targets. Our data indicate that the targeting mechanism specified by CRISPR/Cas9 forces integration into genomic regions that are otherwise poor targets for SB transposition. Future modifications of this technology may allow the development of methods for specific gene insertion for precision genetic engineering.
Collapse
Affiliation(s)
- Adrian Kovač
- Transposition and Genome Engineering, Division of Medical Biotechnology, Paul Ehrlich InstituteLangenGermany
| | - Csaba Miskey
- Transposition and Genome Engineering, Division of Medical Biotechnology, Paul Ehrlich InstituteLangenGermany
| | | | - Esther Grueso
- Transposition and Genome Engineering, Division of Medical Biotechnology, Paul Ehrlich InstituteLangenGermany
| | | | - Zoltán Ivics
- Transposition and Genome Engineering, Division of Medical Biotechnology, Paul Ehrlich InstituteLangenGermany
| |
Collapse
|
267
|
Comprehensive genomic characterization of gene therapy-induced T-cell acute lymphoblastic leukemia. Leukemia 2020; 34:2785-2789. [PMID: 32127638 PMCID: PMC8321895 DOI: 10.1038/s41375-020-0779-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 01/17/2020] [Accepted: 02/18/2020] [Indexed: 02/04/2023]
|
268
|
Gianni F, Belver L, Ferrando A. The Genetics and Mechanisms of T-Cell Acute Lymphoblastic Leukemia. Cold Spring Harb Perspect Med 2020; 10:a035246. [PMID: 31570389 PMCID: PMC7050584 DOI: 10.1101/cshperspect.a035246] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematologic malignancy derived from early T-cell progenitors. The recognition of clinical, genetic, transcriptional, and biological heterogeneity in this disease has already translated into new prognostic biomarkers, improved leukemia animal models, and emerging targeted therapies. This work reviews our current understanding of the molecular mechanisms of T-ALL.
Collapse
Affiliation(s)
- Francesca Gianni
- Institute for Cancer Genetics, Columbia University Medical Center, New York, New York 10032, USA
| | - Laura Belver
- Institute for Cancer Genetics, Columbia University Medical Center, New York, New York 10032, USA
| | - Adolfo Ferrando
- Institute for Cancer Genetics, Columbia University Medical Center, New York, New York 10032, USA
- Department of Pathology, Columbia University Medical Center, New York, New York 10032, USA
- Department of Pediatrics, Columbia University Medical Center, New York, New York 10032, USA
| |
Collapse
|
269
|
Zhang ZY, Thrasher AJ, Zhang F. Gene therapy and genome editing for primary immunodeficiency diseases. Genes Dis 2020; 7:38-51. [PMID: 32181274 PMCID: PMC7063425 DOI: 10.1016/j.gendis.2019.07.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 07/20/2019] [Accepted: 07/22/2019] [Indexed: 12/12/2022] Open
Abstract
In past two decades the gene therapy using genetic modified autologous hematopoietic stem cells (HSCs) transduced with the viral vector has become a promising alternative option for treating primary immunodeficiency diseases (PIDs). Despite of some pitfalls at early stage clinical trials, the field of gene therapy has advanced significantly in the last decade with improvements in viral vector safety, preparatory regime for manufacturing high quality virus, automated CD34 cell purification. Hence, the overall outcome from the clinical trials for the different PIDs has been very encouraging. In addition to the viral vector based gene therapy, the recent fast moving forward developments in genome editing using engineered nucleases in HSCs has provided a new promising platform for the treatment of PIDs. This review provides an overall outcome and progress in gene therapy clinical trials for SCID-X, ADA-SCID, WAS, X- CGD, and the recent developments in genome editing technology applied in HSCs for developing potential therapy, particular in the key studies for PIDs.
Collapse
Affiliation(s)
- Zhi-Yong Zhang
- Department of Immunology and Rheumatology, Children's Hospital of Chongqing Medical University, China
| | - Adrian J. Thrasher
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University Colleage London, UK
| | - Fang Zhang
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University Colleage London, UK
| |
Collapse
|
270
|
Asher DR, Thapa K, Dharia SD, Khan N, Potter RA, Rodino-Klapac LR, Mendell JR. Clinical development on the frontier: gene therapy for duchenne muscular dystrophy. Expert Opin Biol Ther 2020; 20:263-274. [PMID: 32031420 DOI: 10.1080/14712598.2020.1725469] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: The development of adeno-associated virus (AAV) vectors as safe vehicles for in vivo delivery of therapeutic genes has been a major milestone in the advancement of gene therapy, enabling a promising strategy for ameliorating a wide range of diseases, including Duchenne muscular dystrophy (DMD).Areas covered: Based on experience with the development of a gene transfer therapy agent for DMD, we discuss ways in which gene therapy for rare disease challenges traditional clinical development paradigms, and recommend a step-wise approach for design and evaluation to support broader applicability of gene therapy.Expert opinion: The gene therapy development approach should intentionally design the therapeutic construct and the clinical study to systematically evaluate agent delivery, safety, and efficacy. Rigorous preclinical work is essential for establishing an effective gene delivery platform and determining the efficacious dose. Clinical studies should thoroughly evaluate transduction, on-target transgene expression at the tissue and cellular level, and functional efficacy.
Collapse
Affiliation(s)
- Damon R Asher
- Sarepta Therapeutics, Inc, Cambridge, Massachusetts, USA
| | | | - Sachi D Dharia
- Sarepta Therapeutics, Inc, Cambridge, Massachusetts, USA
| | - Navid Khan
- Sarepta Therapeutics, Inc, Cambridge, Massachusetts, USA
| | | | | | - Jerry R Mendell
- Center for Gene Therapy, The Abigail Wexner, Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics and Neurology, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
271
|
Nobles CL, Sherrill-Mix S, Everett JK, Reddy S, Fraietta JA, Porter DL, Frey N, Gill SI, Grupp SA, Maude SL, Siegel DL, Levine BL, June CH, Lacey SF, Melenhorst JJ, Bushman FD. CD19-targeting CAR T cell immunotherapy outcomes correlate with genomic modification by vector integration. J Clin Invest 2020; 130:673-685. [PMID: 31845905 PMCID: PMC6994131 DOI: 10.1172/jci130144] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 10/08/2019] [Indexed: 12/15/2022] Open
Abstract
Chimeric antigen receptor-engineered T cells targeting CD19 (CART19) provide an effective treatment for pediatric acute lymphoblastic leukemia but are less effective for chronic lymphocytic leukemia (CLL), focusing attention on improving efficacy. CART19 harbor an engineered receptor, which is delivered through lentiviral vector integration, thereby marking cell lineages and modifying the cellular genome by insertional mutagenesis. We recently reported that vector integration within the host TET2 gene was associated with CLL remission. Here, we investigated clonal population structure and therapeutic outcomes in another 39 patients by high-throughput sequencing of vector-integration sites. Genes at integration sites enriched in responders were commonly found in cell-signaling and chromatin modification pathways, suggesting that insertional mutagenesis in these genes promoted therapeutic T cell proliferation. We also developed a multivariate model based on integration-site distributions and found that data from preinfusion products forecasted response in CLL successfully in discovery and validation cohorts and, in day 28 samples, reported responders to CLL therapy with high accuracy. These data clarify how insertional mutagenesis can modulate cell proliferation in CART19 therapy and how data on integration-site distributions can be linked to treatment outcomes.
Collapse
MESH Headings
- Antigens, CD19/genetics
- Antigens, CD19/immunology
- Female
- Genetic Vectors
- Humans
- Immunotherapy, Adoptive
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Male
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
Collapse
Affiliation(s)
| | | | | | | | - Joseph A. Fraietta
- Department of Microbiology
- Center for Cellular Immunotherapies
- Department of Pathology and Laboratory Medicine, and
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David L. Porter
- Center for Cellular Immunotherapies
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Division of Oncology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Noelle Frey
- Center for Cellular Immunotherapies
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Saar I. Gill
- Center for Cellular Immunotherapies
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Stephan A. Grupp
- Division of Oncology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Shannon L. Maude
- Division of Oncology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Donald L. Siegel
- Center for Cellular Immunotherapies
- Department of Pathology and Laboratory Medicine, and
| | - Bruce L. Levine
- Center for Cellular Immunotherapies
- Department of Pathology and Laboratory Medicine, and
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Carl H. June
- Center for Cellular Immunotherapies
- Department of Pathology and Laboratory Medicine, and
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Simon F. Lacey
- Center for Cellular Immunotherapies
- Department of Pathology and Laboratory Medicine, and
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - J. Joseph Melenhorst
- Center for Cellular Immunotherapies
- Department of Pathology and Laboratory Medicine, and
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
272
|
Klatt D, Cheng E, Hoffmann D, Santilli G, Thrasher AJ, Brendel C, Schambach A. Differential Transgene Silencing of Myeloid-Specific Promoters in the AAVS1 Safe Harbor Locus of Induced Pluripotent Stem Cell-Derived Myeloid Cells. Hum Gene Ther 2020; 31:199-210. [PMID: 31773990 DOI: 10.1089/hum.2019.194] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Targeted integration into a genomic safe harbor, such as the AAVS1 locus on chromosome 19, promises predictable transgene expression and reduces the risk of insertional mutagenesis in the host genome. The application of gamma-retroviral long terminal repeat (LTR)-driven vectors, which semirandomly integrate into the genome, has previously caused severe adverse events in some clinical studies due to transactivation of neighboring proto-oncogenes. Consequently, the site-specific integration of a therapeutic transgene into a genomic safe harbor locus would allow stable genetic correction with a reduced risk of insertional mutagenesis. However, recent studies revealed that transgene silencing, especially in case of weaker cell type-specific promoters, can occur in the AAVS1 locus of human pluripotent stem cells (PSCs) and can impede transgene expression during differentiation. In this study, we aimed to correct p47phox deficiency, which is the second most common cause of chronic granulomatous disease, by insertion of a therapeutic p47phox transgene into the AAVS1 locus of human induced PSCs (iPSCs) using CRISPR-Cas9. We analyzed transgene expression and functional correction from three different myeloid-specific promoters (miR223, CatG/cFes, and myeloid-related protein 8 [MRP8]). Upon myeloid differentiation of corrected iPSC clones, we observed that the miR223 and CatG/cFes promoters achieved therapeutically relevant levels of p47phox expression and nicotinamide adenine dinucleotide phosphate oxidase activity, whereas the MRP8 promoter was less efficient. Analysis of the different promoters revealed high CpG methylation of the MRP8 promoter in differentiated cells, which correlated with the transgene expression data. In summary, we identified the miR223 and CatG/cFes promoters as cell type-specific promoters that allow stable transgene expression in the AAVS1 locus of iPSC-derived myeloid cells. Our findings further indicate that promoter silencing can occur in the AAVS1 safe harbor locus in differentiated hematopoietic cells and that a comparison of different promoters is necessary to achieve optimal transgene expression for therapeutic application of iPSC-derived cells.
Collapse
Affiliation(s)
- Denise Klatt
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Erica Cheng
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Dirk Hoffmann
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Giorgia Santilli
- Infection, Immunity and Inflammation Program, Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Adrian J Thrasher
- Infection, Immunity and Inflammation Program, Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom.,Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom
| | - Christian Brendel
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
273
|
Retroviral Insertional Mutagenesis in Humans: Evidence for Four Genetic Mechanisms Promoting Expansion of Cell Clones. Mol Ther 2020; 28:352-356. [PMID: 31951833 DOI: 10.1016/j.ymthe.2019.12.009] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/20/2019] [Accepted: 12/21/2019] [Indexed: 01/12/2023] Open
Abstract
Integration of new DNA into a cellular chromosome can alter the activity of nearby genes, sometimes affecting subsequent cell growth. A potent form of insertional mutagenesis involves integration of retroviral DNA produced by reverse transcription, a required step in the replication of retroviruses. In recent years retroviral replication has been adapted to allow new gene addition by retroviral vectors. Early in the history of retrovirus research, analysis of insertional mutagenesis in laboratory animals was found at times to result in transformation, leading to the discovery of cellular proto-oncogenes. In-depth analysis of the genetic consequences showed that integration of retroviral DNA could alter the gene activity in a variety of ways. Mechanisms of retroviral insertional mutagenesis in humans are much less well documented. However, recent work from the gene therapy and HIV fields now specify four genetic mechanisms of retroviral insertional mutagenesis in humans: (1) gene activation by integration of an enhancer sequence encoded in a retroviral vector (enhancer insertion), (2) gene activation by promoter insertion, (3) gene inactivation by insertional disruption, and (4) gene activation by mRNA 3' end substitution. In each example, integration in patients was associated with clonal expansion or frank transformation.
Collapse
|
274
|
Delhove J, Osenk I, Prichard I, Donnelley M. Public Acceptability of Gene Therapy and Gene Editing for Human Use: A Systematic Review. Hum Gene Ther 2020; 31:20-46. [PMID: 31802714 DOI: 10.1089/hum.2019.197] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Gene therapy and gene editing technologies are complex and it can be difficult for the public to understand their possible benefits or side effects. However, patient and public support is critical for the successful adoption of any new technology. Given the recent advances in gene therapy and gene editing, their potential clinical benefits, and the significant attention that has been given to the first-known successful attempt at permanent and heritable changes to the human genome, a systematic review was performed to assess beliefs and attitudes toward gene therapy and gene editing for human use, and to highlight the factors that influence acceptability. A systematic search following Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines was undertaken in April 2018 to identify articles examining opinions and attitudes regarding the acceptability of gene therapy and gene editing. Overall, 1,561 records were retrieved from 4 databases (Ovid Medline, PsycINFO, Scopus, and Web of Science). Duplicates were removed, and titles and abstracts independently screened, leaving 86 full-text articles assessed for eligibility. Following full-text review, 33 were included, with 5 articles added after forward/backward searching. An additional three articles were added following an updated search in March 2019 (total n = 41). Findings from the studies were integrated according to common themes: the impact of demographics; risks versus benefits of success; treatment specifics (e.g., medical vs. other reasons; disease severity and status; somatic vs. germ line; and mode of delivery); moral or ethical issues; and changes with time. In general, perceptions were positive, particularly for medical reasons and fatal diseases, but were also influenced by perceived risk. Somatic therapies had higher levels of acceptability than germ line therapies. While available in various forms, limitations exist in the measurement of perceptions of gene therapy and gene editing. Treatment acceptability is essential for future clinical trials, so it is important for scientists and clinicians to be clear about the risks and benefits of these technologies, and how these are communicated to the public, while encouraging education about genetic therapies to a broad range of individuals.
Collapse
Affiliation(s)
- Juliette Delhove
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, Australia
- Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, Australia
| | - Ivana Osenk
- College of Nursing and Health Sciences, Flinders University, Bedford Park, Australia
| | - Ivanka Prichard
- College of Nursing and Health Sciences, Flinders University, Bedford Park, Australia
| | - Martin Donnelley
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, Australia
- Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, Australia
| |
Collapse
|
275
|
Han J, Gao F, Geng S, Ye X, Wang T, Du P, Cai Z, Fu Z, Zhao Z, Shi L, Li Q, Cai J. Minicircle DNA-Engineered CAR T Cells Suppressed Tumor Growth in Mice. Mol Cancer Ther 2020; 19:178-186. [PMID: 31582530 DOI: 10.1158/1535-7163.mct-19-0204] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 07/04/2019] [Accepted: 09/27/2019] [Indexed: 11/16/2022]
Abstract
Viral-based chimeric antigen receptor-engineered T (CAR T)-cell manufacturing has potential safety risks and relatively high costs. The nonviral minicircle DNA (mcDNA) is safer for patients, cheaper to produce, and may be a more suitable technique to generate CAR T cells. In this study, we produced mcDNA-based CAR T cells specifically targeting prostate stem cell antigen (PSCA; mcDNA-PSCA-CAR T cells). Our results showed that mcDNA-PSCA-CAR T cells persisted in mouse peripheral blood as long as 28 days and demonstrated more CAR T-cell infiltration, higher cytokine secretion levels, and better antitumor effects. Together, our results suggest that mcDNA-CAR can be a safe and cost-effective platform to produce CAR T cells.
Collapse
Affiliation(s)
- Jinsheng Han
- Department of Surgery, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Fei Gao
- Department of Surgery, Hebei Medical University, Shijiazhuang, Hebei, China
- Department of Surgery and Oncology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Songsong Geng
- Hebei Engineering Technology Research Center for Cell Therapy, Hebei HOFOY Biotech Company Ltd., Shijiazhuang, Hebei, China
| | - Xueshuai Ye
- Department of Surgery, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Engineering Technology Research Center for Cell Therapy, Hebei HOFOY Biotech Company Ltd., Shijiazhuang, Hebei, China
| | - Tie Wang
- Department of Surgery, Hebei Cangzhou Hospital of Integrated Traditional Chinese and Western Medicine, Cangzhou, Hebei, China
| | - Pingping Du
- Hebei Engineering Technology Research Center for Cell Therapy, Hebei HOFOY Biotech Company Ltd., Shijiazhuang, Hebei, China
| | - Ziqi Cai
- Hebei Engineering Technology Research Center for Cell Therapy, Hebei HOFOY Biotech Company Ltd., Shijiazhuang, Hebei, China
| | - Zexian Fu
- Hebei Engineering Technology Research Center for Cell Therapy, Hebei HOFOY Biotech Company Ltd., Shijiazhuang, Hebei, China
- Department of Surgery, Affiliated Hospital of Hebei Engineering University, Handan, Hebei, China
| | - Zhilong Zhao
- Hebei Engineering Technology Research Center for Cell Therapy, Hebei HOFOY Biotech Company Ltd., Shijiazhuang, Hebei, China
- Department of Surgery, the Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Long Shi
- Hebei Engineering Technology Research Center for Cell Therapy, Hebei HOFOY Biotech Company Ltd., Shijiazhuang, Hebei, China
- Department of Oncology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Qingxia Li
- Department of Surgery and Oncology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Jianhui Cai
- Department of Surgery, Hebei Medical University, Shijiazhuang, Hebei, China.
- Department of Surgery and Oncology, Hebei General Hospital, Shijiazhuang, Hebei, China
- Hebei Engineering Technology Research Center for Cell Therapy, Hebei HOFOY Biotech Company Ltd., Shijiazhuang, Hebei, China
| |
Collapse
|
276
|
Pai SY. Treatment of primary immunodeficiency with allogeneic transplant and gene therapy. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2019; 2019:457-465. [PMID: 31808905 PMCID: PMC6913427 DOI: 10.1182/hematology.2019000052] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
The treatment of primary immunodeficiency disorders with allogeneic hematopoietic cell transplantation (HCT) has a history dating back to 1968 with the first successful transplant for a patient with severe combined immunodeficiency (SCID). The omission of conditioning for patients with SCID owing to their inability to reject allogeneic grafts and the increasing use of reduced intensity conditioning regimens often result in a state of mixed or split donor-recipient chimerism. The use of gene therapy (GT) via retroviral or lentiviral transduction of autologous CD34+ hematopoietic stem and progenitor cells is expected to correct only a portion of the hematopoietic stem cell compartment. The consequences of partial correction after either form of cellular therapy differ according to how the genetic deficiency affects immune cell development and function. Moreover, the conditioning regimen or lack thereof impacts the cell lineages at risk of partial correction. Advances in our understanding of immune reconstitution after HCT and GT for SCID, Wiskott-Aldrich syndrome, and chronic granulomatous disease are discussed.
Collapse
Affiliation(s)
- Sung-Yun Pai
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, MA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA; and
- Harvard Medical School, Boston, MA
| |
Collapse
|
277
|
Li C, Lieber A. Adenovirus vectors in hematopoietic stem cell genome editing. FEBS Lett 2019; 593:3623-3648. [PMID: 31705806 PMCID: PMC10473235 DOI: 10.1002/1873-3468.13668] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 10/23/2019] [Accepted: 10/27/2019] [Indexed: 12/13/2022]
Abstract
Genome editing of hematopoietic stem cells (HSCs) represents a therapeutic option for a number of hematological genetic diseases, as HSCs have the potential for self-renewal and differentiation into all blood cell lineages. This review presents advances of genome editing in HSCs utilizing adenovirus vectors as delivery vehicles. We focus on capsid-modified, helper-dependent adenovirus vectors that are devoid of all viral genes and therefore exhibit an improved safety profile. We discuss HSC genome engineering for several inherited disorders and infectious diseases including hemoglobinopathies, Fanconi anemia, hemophilia, and HIV-1 infection by ex vivo and in vivo editing in transgenic mice, nonhuman primates, as well as in human CD34+ cells. Mechanisms of therapeutic gene transfer including episomal expression of designer nucleases and base editors, transposase-mediated random integration, and targeted homology-directed repair triggered integration into selected genomic safe harbor loci are also reviewed.
Collapse
Affiliation(s)
- Chang Li
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA, USA
| | - André Lieber
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA, USA
- Department of Pathology, University of Washington, Seattle, WA, USA
| |
Collapse
|
278
|
Koniaeva E, Stahlhut M, Lange L, Sauer MG, Kustikova OS, Schambach A. Conditional Immortalization of Lymphoid Progenitors via Tetracycline-Regulated LMO2 Expression. Hum Gene Ther 2019; 31:183-198. [PMID: 31760808 DOI: 10.1089/hum.2019.212] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Conditional immortalization of hematopoietic progenitors through lentiviral expression of selected transcription factors in hematopoietic stem and progenitor cells provides a promising tool to study stem cell and leukemia biology. In this study, to generate conditionally immortalized lymphoid progenitor (ciLP) cell lines, murine hematopoietic progenitor cells were transduced with an inducible lentiviral "all-in-one" vector expressing LMO2 under doxycycline (DOX) stimulation and the reverse tetracycline-regulated transactivator (rtTA3). For selection of LMO2-expressing ciLPs (LMO2-ciLPs) and longitudinal manipulation in T cell differentiation lymphoid conditions, we developed a robust approach based on coculture with OP9-DL1 stromal cells and improved cytokine conditions allowing a controlled balance between cell proliferation and differentiation in vitro. LMO2-ciLP cell lines with the highest proliferation, vector copy number, and similar insertion pattern were selected for LMO2 "on/off" in vitro study. LMO2 expression under DOX induction resulted in a double negative (DN) 2 differentiation arrest and a propagation of CD44+CD25- myeloid cell population characterized by lymphoid and myeloid phenotypes, respectively. Both DN2 and CD44+CD25- myeloid cell subpopulations expressed c-KIT, suggesting that LMO2-ciLPs were similar to uncommitted progenitors under DOX supplementation. DOX removal resulted in cessation of ectopic LMO2 expression and LMO2-ciLPs continued T cell lymphoid differentiation accompanied by c-KIT downregulation and interleukin 7 receptor expression. Switching off LMO2 expression was accompanied by increased Notch signaling and significant reduction of the CD44+CD25- myeloid cell population under T cell differentiation lymphoid conditions. Although vector insertions in cooperation with LMO2 expression could influence the fate of LMO2-ciLPs and additional experiments are required to evaluate it, our approach provides a promising tool to investigate mechanisms underlying stem cell, leukemia, and lymphocyte biology, leading to novel approaches for disease modeling and therapy evaluation.
Collapse
Affiliation(s)
- Ekaterina Koniaeva
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,Cluster of Excellence REBIRTH, Hannover Medical School, Hannover, Germany
| | - Maike Stahlhut
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,Cluster of Excellence REBIRTH, Hannover Medical School, Hannover, Germany
| | - Lucas Lange
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,Cluster of Excellence REBIRTH, Hannover Medical School, Hannover, Germany
| | - Martin G Sauer
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Olga S Kustikova
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,Cluster of Excellence REBIRTH, Hannover Medical School, Hannover, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,Cluster of Excellence REBIRTH, Hannover Medical School, Hannover, Germany.,Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
279
|
Bari S, Chong P, Hwang WYK. Expansion of Haematopoietic Stem and Progenitor Cells: Paving the Way for Next-Generation Haematopoietic Stem Cell Transplantation. BLOOD CELL THERAPY 2019; 2:58-67. [PMID: 37588101 PMCID: PMC10427230 DOI: 10.31547/bct-2019-004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 09/18/2019] [Indexed: 08/18/2023]
Abstract
Haematopoietic stem cell transplantation (HSCT) is now an established practice with over 70,000 transplants performed annually, and over 1.5 million around the world so far. The practice of HSCT has improved over the years due to advances in conditioning regiments, preparatory practices for patients leading up to the transplant, graft versus host disease (GVHD) and infection prophylaxis, as well as a better selection of patients. However, in many instances, the stem cells supplied to the patient may not be adequate for optimal transplantation outcomes. This may be seen in a few areas including umbilical cord blood transplantation, inadequate bone marrow, peripheral blood stem cell harvest, or gene therapy. Growing and expanding HSCs in culture would provide an increase in cell numbers prior to stem cell infusion and accelerate haematopoietic recovery, resulting in improved outcomes. Several new technologies have emerged in recent years, which have facilitated the expansion of haematopoietic stem and progenitor cells (HSPCs) in culture with good outcomes in vitro, in vivo, and in clinical trials. In this review, we will outline some of the reasons for the expansion of HSPCs as well as the new technologies facilitating the advances in HSCT.
Collapse
Affiliation(s)
- Sudipto Bari
- National Cancer Centre Singapore
- Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore
| | | | - William Ying Khee Hwang
- National Cancer Centre Singapore
- Department of Haematology, Singapore General Hospital, Singapore
- Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore
| |
Collapse
|
280
|
Ishii T, Beriain IDM. Safety of Germline Genome Editing for Genetically Related "Future" Children as Perceived by Parents. CRISPR J 2019; 2:370-375. [PMID: 31746634 PMCID: PMC6919246 DOI: 10.1089/crispr.2019.0010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The social acceptability of germline genome editing (GGE) depends on its perceived safety, as well as respect for reproductive autonomy. However, it is doubtful that prospective parents sufficiently understand the risks of GGE. In the future, the use of GGE in specific situations seems plausible, as it offers couples potential means to safeguard genetically related future children from a serious disease and overcome infertility due to a gene mutation. Should GGE fail, however, some couples may be obliged to abort affected fetuses, or give birth to adversely affected children, which would be a tragedy. Some children might develop diseases later in life due to overlooked off-target mutations. Compounding this, some parents are unlikely to inform their offspring about the details of conception, hampering necessary follow-up. Prospective parents, scientists and policy makers should carefully discuss the safety implications of GGE for genetically related future children.
Collapse
Affiliation(s)
- Tetsuya Ishii
- Office of Health and Safety, Hokkaido University, Sapporo, Japan
| | - Iñigo de Miguel Beriain
- Law and the Human Genome RG, University of the Basque Country UPV/EHU, Faculty of Law, Leioa, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
281
|
Moço PD, Aharony N, Kamen A. Adeno-Associated Viral Vectors for Homology-Directed Generation of CAR-T Cells. Biotechnol J 2019; 15:e1900286. [PMID: 31642193 DOI: 10.1002/biot.201900286] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 10/06/2019] [Indexed: 12/19/2022]
Abstract
Immunotherapy with T cells expressing chimeric antigen receptors (CAR) is an emerging and promising treatment against refractory cancers. However, the currently adopted methods of modification of T cells pose a risk of insertional oncogenesis because lentiviral and retroviral vectors integrate the CAR transgene in a semi-random fashion. In addition, this therapy is only available using autologous cells, which create problems in production and limit the access for patients who have their T cells depleted. One modification method that shows the ability to overcome both drawbacks is the knock-in of the CAR simultaneously knocking-out genes that prevent allogeneic therapy, such as the endogenous T cell receptor. In this mini-review, the authors present recent efforts to develop safer universal CAR-T cells. More specifically, the combined application of target-directed nucleases, which create a double-strand break at a specific genome locus, and the delivery of CAR DNA via adeno-associated viral vectors for subsequent integration via homologous recombination and silencing of the targeted gene is focused on.
Collapse
Affiliation(s)
- Pablo D Moço
- Department of Bioengineering, McGill University, Montréal, H3A 0E9, Canada
| | - Noga Aharony
- Department of Bioengineering, McGill University, Montréal, H3A 0E9, Canada
| | - Amine Kamen
- Department of Bioengineering, McGill University, Montréal, H3A 0E9, Canada
| |
Collapse
|
282
|
Hematopoietic stem cell gene therapy: The optimal use of lentivirus and gene editing approaches. Blood Rev 2019; 40:100641. [PMID: 31761379 DOI: 10.1016/j.blre.2019.100641] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/06/2019] [Accepted: 11/11/2019] [Indexed: 12/26/2022]
Abstract
Due to pioneering in vitro investigations on gene modification, gene engineering platforms have incredibly improved to a safer and more powerful tool for the treatment of multiple blood and immune disorders. Likewise, several clinical trials have been initiated combining autologous hematopoietic stem cell transplantation (auto-HSCT) with gene therapy (GT) tools. As several GT modalities such as lentivirus and gene editing tools have a long developmental path ahead to diminish its negative side effects, it is hard to decide which modality is optimal for treating a specific disease. Gene transfer by lentiviruses is the platform of choice for loss-of-mutation diseases, whereas gene correction/addition or gene disruption by gene editing tools, mainly CRISPR/Cas9, is likely to be more efficient in diseases where tight regulation is needed. Therefore, in this review, we compiled pertinent information about lentiviral gene transfer and CRISPR/Cas9 gene editing, their evolution to a safer platform for HSCT, and their applications on other types of gene disorders based on the etiology of the disease and cell fitness.
Collapse
|
283
|
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: 25] [Impact Index Per Article: 4.2] [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
|
284
|
Radtke S, Humbert O, Kiem HP. Mouse models in hematopoietic stem cell gene therapy and genome editing. Biochem Pharmacol 2019; 174:113692. [PMID: 31705854 DOI: 10.1016/j.bcp.2019.113692] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 11/01/2019] [Indexed: 12/26/2022]
Abstract
Gene therapy has become an important treatment option for a variety of hematological diseases. The biggest advances have been made with CAR T cells and many of those studies are now FDA approved as a routine treatment for some hematologic malignancies. Hematopoietic stem cell (HSC) gene therapy is not far behind with treatment approvals granted for beta-hemoglobinopathies and adenosine deaminase severe combined immune deficiency (ADA-SCID), and additional approbations currently being sought. With the current pace of research, the significant investment of biotech companies, and the continuously growing toolbox of viral as well as non-viral gene delivery methods, the development of new ex vivo and in vivo gene therapy approaches is at an all-time high. Research in the field of gene therapy has been ongoing for more than 4 decades with big success stories as well as devastating drawbacks along the way. In particular, the damaging effect of uncontrolled viral vector integration observed in the initial gene therapy applications in the 90s led to a more comprehensive upfront safety assessment of treatment strategies. Since the late 90s, an important read-out to comprehensively assess the quality and safety of cell products has come forward with the mouse xenograft model. Here, we review the use of mouse models across the different stages of basic, pre-clinical and translational research towards the clinical application of HSC-mediated gene therapy and editing approaches.
Collapse
Affiliation(s)
- Stefan Radtke
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | - Olivier Humbert
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | - Hans-Peter Kiem
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA; Department of Pathology, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
285
|
Feinberg D, Paul B, Kang Y. The promise of chimeric antigen receptor (CAR) T cell therapy in multiple myeloma. Cell Immunol 2019; 345:103964. [PMID: 31492448 PMCID: PMC6832886 DOI: 10.1016/j.cellimm.2019.103964] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/08/2019] [Accepted: 08/10/2019] [Indexed: 12/19/2022]
Abstract
A cure for multiple myeloma (MM), a malignancy of plasma cells, remains elusive. Nearly all myeloma patients will eventually relapse and develop resistance to currently available treatments. There is an unmet medical need to develop novel and effective therapies that can induce sustained responses. Early phase clinical trials using chimeric antigen receptor (CAR) T cell therapy have shown great promise in the treatment of relapsed and/or refractory MM. In this review article, we provide an overview of the CAR constructs, the gene transfer vector systems, and approaches for T cell activation and expansion. We then summarize the outcomes of several early phase clinical trials of CAR T cell therapy in MM and the novel CAR T targets that are under development. Finally, we explore the potential mechanisms that result in disease relapse after CAR T therapy and propose future directions in CAR T therapy in MM.
Collapse
MESH Headings
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Hematopoietic Stem Cell Transplantation/methods
- Humans
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/trends
- Multiple Myeloma/immunology
- Multiple Myeloma/metabolism
- Multiple Myeloma/therapy
- Neoplasm Recurrence, Local
- Outcome Assessment, Health Care
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Daniel Feinberg
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC 27710, USA
| | - Barry Paul
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC 27710, USA
| | - Yubin Kang
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
286
|
Eichstadt S, Barriga M, Ponakala A, Teng C, Nguyen NT, Siprashvili Z, Nazaroff J, Gorell ES, Chiou AS, Taylor L, Khuu P, Keene DR, Rieger K, Khosla RK, Furukawa LK, Lorenz HP, Marinkovich MP, Tang JY. Phase 1/2a clinical trial of gene-corrected autologous cell therapy for recessive dystrophic epidermolysis bullosa. JCI Insight 2019; 4:130554. [PMID: 31578311 DOI: 10.1172/jci.insight.130554] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 08/31/2019] [Indexed: 01/08/2023] Open
Abstract
BACKGROUNDRecessive dystrophic epidermolysis bullosa (RDEB) patients have mutations in the COL7A1 gene and thus lack functional type VII collagen (C7) protein; they have marked skin fragility and blistering. This single-center phase 1/2a open-label study evaluated the long-term efficacy, safety, and patient-reported outcomes in RDEB patients treated with gene-corrected autologous cell therapy.METHODSAutologous keratinocytes were isolated from participant skin biopsies. Epidermal sheets were prepared from cells transduced with a retrovirus carrying the full-length human COL7A1 gene. These gene-corrected autologous epidermal sheets measured 5 × 7 cm (35 cm2) and were transplanted onto 6 wound sites in each of 7 adult participants (n = 42 sites total) from 2013 to 2017. Participants were followed for 2 to 5 years.RESULTSNo participants experienced any serious related adverse events. Wound healing of 50% or greater by Investigator Global Assessment was present in 95% (36 of 38) of treated wounds versus 0% (0 of 6) of untreated control wounds at 6 months (P < 0.0001). At year 1, 68% (26 of 38) of treated wounds had 50% or greater healing compared with 17% (1 of 6) of control wounds (P = 0.025). At year 2, 71% (27 of 38) of treated wounds had 50% or greater healing compared with 17% (1 of 6) of control wounds (P = 0.019).CONCLUSIONC7 expression persisted up to 2 years after treatment in 2 participants. Treated wounds with 50% or greater healing demonstrated improvement in patient-reported pain, itch, and wound durability. This study provides additional data to support the clinically meaningful benefit of treating chronic RDEB wounds with ex vivo, C7 gene-corrected autologous cell therapy. This approach was safe and promoted wound healing that was associated with improved patient-reported outcomes.TRIAL REGISTRATIONClinicaltrials.gov identifier: NCT01263379.FUNDINGEpidermolysis Bullosa Research Partnership, Epidermolysis Bullosa Medical Research Foundation, NIH R01 AR055914, Office of Research and Development at the Palo Alto Veteran's Affairs Medical Center, and the Dermatology Foundation.
Collapse
Affiliation(s)
- Shaundra Eichstadt
- Stanford University School of Medicine, Department of Dermatology, Redwood City, California, USA
| | - Melissa Barriga
- Stanford University School of Medicine, Department of Dermatology, Redwood City, California, USA
| | - Anusha Ponakala
- Stanford University School of Medicine, Department of Dermatology, Redwood City, California, USA
| | - Claudia Teng
- Stanford University School of Medicine, Department of Dermatology, Redwood City, California, USA
| | | | - Zurab Siprashvili
- Stanford University School of Medicine, Department of Dermatology, Redwood City, California, USA
| | - Jaron Nazaroff
- Stanford University School of Medicine, Department of Dermatology, Redwood City, California, USA
| | - Emily S Gorell
- Stanford University School of Medicine, Department of Dermatology, Redwood City, California, USA
| | - Albert S Chiou
- Stanford University School of Medicine, Department of Dermatology, Redwood City, California, USA
| | - Lisa Taylor
- Lucile Packard Children's Hospital, Stanford Children's Health, Stanford, California, USA
| | - Phuong Khuu
- Lucile Packard Children's Hospital, Stanford Children's Health, Stanford, California, USA
| | | | - Kerri Rieger
- Stanford University School of Medicine, Department of Dermatology, Redwood City, California, USA
| | - Rohit K Khosla
- Stanford University School of Medicine, Department of Dermatology, Redwood City, California, USA
| | - Louise K Furukawa
- Lucile Packard Children's Hospital, Stanford Children's Health, Stanford, California, USA
| | - H Peter Lorenz
- Lucile Packard Children's Hospital, Stanford Children's Health, Stanford, California, USA
| | - M Peter Marinkovich
- Stanford University School of Medicine, Department of Dermatology, Redwood City, California, USA.,Veterans Affairs Medical Center, Palo Alto, California, USA
| | - Jean Y Tang
- Stanford University School of Medicine, Department of Dermatology, Redwood City, California, USA
| |
Collapse
|
287
|
Wünsche P, Eckert ESP, Holland-Letz T, Paruzynski A, Hotz-Wagenblatt A, Fronza R, Rath T, Gil-Farina I, Schmidt M, von Kalle C, Klein C, Ball CR, Herbst F, Glimm H. Mapping Active Gene-Regulatory Regions in Human Repopulating Long-Term HSCs. Cell Stem Cell 2019; 23:132-146.e9. [PMID: 29979988 DOI: 10.1016/j.stem.2018.06.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 04/03/2018] [Accepted: 06/08/2018] [Indexed: 12/29/2022]
Abstract
Genes that regulate hematopoietic stem cell (HSC) self-renewal, proliferation, and differentiation are tightly controlled by regulatory regions. However, mapping such regions relies on surface markers and immunophenotypic definition of HSCs. Here, we use γ-retroviral integration sites (γRV ISs) from a gene therapy trial for 10 patients with Wiskott-Aldrich syndrome to mark active enhancers and promoters in functionally defined long-term repopulating HSCs. Integration site clusters showed the highest ATAC-seq signals at HSC-specific peaks and strongly correlated with hematopoietic risk variants. Tagged genes were significantly enriched for HSC gene sets. We were able to map over 3,000 HSC regulatory regions in late-contributing HSCs, and we used these data to identify miR-10a and miR-335 as two miRNAs regulating early hematopoiesis. In this study, we show that viral insertion sites can be used as molecular tags to assess chromatin conformation on functionally defined cell populations, thereby providing a genome-wide resource for regulatory regions in human repopulating long-term HSCs.
Collapse
Affiliation(s)
- Peer Wünsche
- Department of Translational and Functional Cancer Genomics, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Elias S P Eckert
- Department of Translational and Functional Cancer Genomics, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Tim Holland-Letz
- Department of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Agnes Hotz-Wagenblatt
- Core Facility Omics IT and Data Management, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Raffaele Fronza
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tim Rath
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Irene Gil-Farina
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Manfred Schmidt
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany; GeneWerk GmbH, Heidelberg, Germany
| | - Christof von Kalle
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christoph Klein
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, Germany
| | - Claudia R Ball
- Department of Translational Medical Oncology, NCT-Dresden, University Hospital, Carl Gustav Carus, Technische Universität Dresden, Dresden and DKFZ, Heidelberg, Germany
| | - Friederike Herbst
- Department of Translational and Functional Cancer Genomics, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hanno Glimm
- Department of Translational and Functional Cancer Genomics, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Translational Medical Oncology, NCT-Dresden, University Hospital, Carl Gustav Carus, Technische Universität Dresden, Dresden and DKFZ, Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany.
| |
Collapse
|
288
|
Gollomp KL, Doshi BS, Arruda VR. Gene therapy for hemophilia: Progress to date and challenges moving forward. Transfus Apher Sci 2019; 58:602-612. [DOI: 10.1016/j.transci.2019.08.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
289
|
Karponi G, Zogas N. Gene Therapy For Beta-Thalassemia: Updated Perspectives. APPLICATION OF CLINICAL GENETICS 2019; 12:167-180. [PMID: 31576160 PMCID: PMC6765258 DOI: 10.2147/tacg.s178546] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 09/11/2019] [Indexed: 12/26/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation was until very recently, the only permanent curative option available for patients suffering from transfusion-dependent beta-thalassemia. Gene therapy, by autologous transplantation of genetically modified hematopoietic stem cells, currently represents a novel therapeutic promise, after many years of extensive preclinical research for the optimization of gene transfer protocols. Nowadays, clinical trials being held on a worldwide setting, have demonstrated that, by re-establishing effective hemoglobin production, patients may be rendered transfusion- and chelation-independent and evade the immunological complications that normally accompany allogeneic hematopoietic stem cell transplantation. The present review will offer a retrospective scope of the long way paved towards successful implementation of gene therapy for beta-thalassemia, and will pinpoint the latest strategies employed to increase globin expression that extend beyond the classic transgene addition perspective. A thorough search was performed using Pubmed in order to identify studies that provide a proof of principle on the aforementioned topic at a preclinical and clinical level. Inclusion criteria also regarded gene transfer technologies of the past two decades, as well as publications outlining the pitfalls that precluded earlier successful implementation of gene therapy for beta-thalassemia. Overall, after decades of research, that included both successes and pitfalls, the path towards a permanent, donor-irrespective cure for beta-thalassemia patients is steadily becoming a realistic approach.
Collapse
Affiliation(s)
- Garyfalia Karponi
- Department of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos Zogas
- Department of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
290
|
Pouzolles M, Machado A, Guilbaud M, Irla M, Gailhac S, Barennes P, Cesana D, Calabria A, Benedicenti F, Sergé A, Raman I, Li QZ, Montini E, Klatzmann D, Adjali O, Taylor N, Zimmermann VS. Intrathymic adeno-associated virus gene transfer rapidly restores thymic function and long-term persistence of gene-corrected T cells. J Allergy Clin Immunol 2019; 145:679-697.e5. [PMID: 31513879 DOI: 10.1016/j.jaci.2019.08.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 07/28/2019] [Accepted: 08/05/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND Patients with T-cell immunodeficiencies are generally treated with allogeneic hematopoietic stem cell transplantation, but alternatives are needed for patients without matched donors. An innovative intrathymic gene therapy approach that directly targets the thymus might improve outcomes. OBJECTIVE We sought to determine the efficacy of intrathymic adeno-associated virus (AAV) serotypes to transduce thymocyte subsets and correct the T-cell immunodeficiency in a zeta-associated protein of 70 kDa (ZAP-70)-deficient murine model. METHODS AAV serotypes were injected intrathymically into wild-type mice, and gene transfer efficiency was monitored. ZAP-70-/- mice were intrathymically injected with an AAV8 vector harboring the ZAP70 gene. Thymus structure, immunophenotyping, T-cell receptor clonotypes, T-cell function, immune responses to transgenes and autoantibodies, vector copy number, and integration were evaluated. RESULTS AAV8, AAV9, and AAV10 serotypes all transduced thymocyte subsets after in situ gene transfer, with transduction of up to 5% of cells. Intrathymic injection of an AAV8-ZAP-70 vector into ZAP-70-/- mice resulted in a rapid thymocyte differentiation associated with the development of a thymic medulla. Strikingly, medullary thymic epithelial cells expressing the autoimmune regulator were detected within 10 days of gene transfer, correlating with the presence of functional effector and regulatory T-cell subsets with diverse T-cell receptor clonotypes in the periphery. Although thymocyte reconstitution was transient, gene-corrected peripheral T cells harboring approximately 1 AAV genome per cell persisted for more than 40 weeks, and AAV vector integration was detected. CONCLUSIONS Intrathymic AAV-transduced progenitors promote a rapid restoration of the thymic architecture, with a single wave of thymopoiesis generating long-term peripheral T-cell function.
Collapse
Affiliation(s)
- Marie Pouzolles
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Alice Machado
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Mickaël Guilbaud
- INSERM UMR1089, Université de Nantes, Centre Hospitalier Universitaire de Nantes, Nantes, France
| | - Magali Irla
- Center of Immunology Marseille-Luminy (CIML), INSERM U1104, CNRS UMR7280, Aix-Marseille Université UM2, Marseille, France
| | - Sarah Gailhac
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Pierre Barennes
- Sorbonne Université, INSERM, Immunology-Immunopathology-Immunotherapy (i3), Paris, France
| | - Daniela Cesana
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS, San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Calabria
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS, San Raffaele Scientific Institute, Milan, Italy
| | - Fabrizio Benedicenti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS, San Raffaele Scientific Institute, Milan, Italy
| | - Arnauld Sergé
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Indu Raman
- Microarray Core Facility, University of Texas Southwestern Medical Center, Dallas, Tex
| | - Quan-Zhen Li
- Microarray Core Facility, University of Texas Southwestern Medical Center, Dallas, Tex; Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Tex
| | - Eugenio Montini
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS, San Raffaele Scientific Institute, Milan, Italy
| | - David Klatzmann
- Sorbonne Université, INSERM, Immunology-Immunopathology-Immunotherapy (i3), Paris, France; AP-HP, Hôpital Pitié-Salpêtrière, Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (i2B), Paris, France
| | - Oumeya Adjali
- INSERM UMR1089, Université de Nantes, Centre Hospitalier Universitaire de Nantes, Nantes, France.
| | - Naomi Taylor
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France; Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md.
| | - Valérie S Zimmermann
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France.
| |
Collapse
|
291
|
Balakrishnan B, David E. Biopolymers augment viral vectors based gene delivery. J Biosci 2019; 44:84. [PMID: 31502562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The success of viral vectors mediated gene therapy is still hampered by immunogenicity and insufficient transgene expression. Alternatively, non-viral vectors mediated gene delivery has the advantage of low immunogenicity despite showing low transgene expression. By carefully considering the advantages of each approach, hybrid vectors are currently being developed by modifying the viral vectors using non-viral biopolymers. This review provides an overview of the hybrid vectors currently being developed.
Collapse
Affiliation(s)
- Balaji Balakrishnan
- Department of Haematology, Christian Medical College, Vellore 632004, Tamil Nadu, India
| | | |
Collapse
|
292
|
McBride DA, Kerr MD, Wai SL, Shah NJ. Applications of molecular engineering in T-cell-based immunotherapies. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 11:e1557. [PMID: 30972976 PMCID: PMC7869905 DOI: 10.1002/wnan.1557] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/24/2019] [Accepted: 03/11/2019] [Indexed: 02/06/2023]
Abstract
Harnessing an individual's immune cells to mediate antitumor and antiviral responses is a life-saving option for some patients with otherwise intractable forms of cancer and infectious disease. In particular, T-cell-based engineered immune cells are a powerful new class of therapeutics with remarkable efficacy. Clinical experience has helped to define some of the major challenges for reliable, safe, and effective deployment of T-cells against a broad range of diseases. While poised to revolutionize immunotherapy, scalable manufacturing, safety, specificity, and the development of resistance are potential roadblocks in their widespread usage. The development of molecular engineering tools to allow for the direct or indirect engineering of T-cells to enable one to troubleshoot delivery issues, amplify immunomodulatory effects, integrate the synergistic effects of different molecules, and home to the target cells in vivo. In this review, we will analyze thus-far developed cell- and material-based tools for enhancing T-cell therapies, including methods to improve safety and specificity, enhancing efficacy, and overcoming limitations in scalable manufacturing. We summarize the potential of T-cells as immune modulating therapies and the potential future directions for enabling their adoption for a broad range of diseases. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Nanotechnology Approaches to Biology > Cells at the Nanoscale.
Collapse
Affiliation(s)
- David A McBride
- Department of Nanoengineering, University of California, San Diego, California
- Program in Chemical Engineering, University of California, San Diego, California
- Center for Nano-Immuno Engineering, University of California, San Diego, California
| | - Matthew D Kerr
- Department of Nanoengineering, University of California, San Diego, California
- Program in Chemical Engineering, University of California, San Diego, California
- Center for Nano-Immuno Engineering, University of California, San Diego, California
| | - Shinya L Wai
- Department of Nanoengineering, University of California, San Diego, California
- Center for Nano-Immuno Engineering, University of California, San Diego, California
| | - Nisarg J Shah
- Department of Nanoengineering, University of California, San Diego, California
- Program in Chemical Engineering, University of California, San Diego, California
- Center for Nano-Immuno Engineering, University of California, San Diego, California
- Graduate Program in Immunology, University of California, San Diego, California
- San Diego Center for Precision Immunotherapy, University of California, San Diego, California
| |
Collapse
|
293
|
|
294
|
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
|
295
|
Affiliation(s)
- Katherine A High
- From Spark Therapeutics, Philadelphia (K.A.H.); and Stanford University, Stanford, CA (M.G.R.)
| | - Maria G Roncarolo
- From Spark Therapeutics, Philadelphia (K.A.H.); and Stanford University, Stanford, CA (M.G.R.)
| |
Collapse
|
296
|
LMO2 activation by deacetylation is indispensable for hematopoiesis and T-ALL leukemogenesis. Blood 2019; 134:1159-1175. [PMID: 31366618 DOI: 10.1182/blood.2019000095] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 07/01/2019] [Indexed: 12/19/2022] Open
Abstract
Hematopoietic transcription factor LIM domain only 2 (LMO2), a member of the TAL1 transcriptional complex, plays an essential role during early hematopoiesis and is frequently activated in T-cell acute lymphoblastic leukemia (T-ALL) patients. Here, we demonstrate that LMO2 is activated by deacetylation on lysine 74 and 78 via the nicotinamide phosphoribosyltransferase (NAMPT)/sirtuin 2 (SIRT2) pathway. LMO2 deacetylation enables LMO2 to interact with LIM domain binding 1 and activate the TAL1 complex. NAMPT/SIRT2-mediated activation of LMO2 by deacetylation appears to be important for hematopoietic differentiation of induced pluripotent stem cells and blood formation in zebrafish embryos. In T-ALL, deacetylated LMO2 induces expression of TAL1 complex target genes HHEX and NKX3.1 as well as LMO2 autoregulation. Consistent with this, inhibition of NAMPT or SIRT2 suppressed the in vitro growth and in vivo engraftment of T-ALL cells via diminished LMO2 deacetylation. This new molecular mechanism may provide new therapeutic possibilities in T-ALL and may contribute to the development of new methods for in vitro generation of blood cells.
Collapse
|
297
|
Raboso-Gallego J, Casado-García A, Isidro-Hernández M, Vicente-Dueñas C. Epigenetic Priming in Childhood Acute Lymphoblastic Leukemia. Front Cell Dev Biol 2019; 7:137. [PMID: 31380372 PMCID: PMC6652134 DOI: 10.3389/fcell.2019.00137] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 07/05/2019] [Indexed: 01/28/2023] Open
Abstract
Leukemogenesis is considered to be a process by which a normal cell acquires new but aberrant identity in order to disseminate a malignant clonal population. Under this setting, the phenotype of the leukemic cells is identical to the leukemia-initiating cell in which the genetic insult is taking place. Thus, with some exceptions, B-cell and T-cell childhood leukemias are supposed to arise from B- or T-committed cells. In contrast, several recent studies have revealed that genetic alterations may act in a “hit-and-run” way in the cell-of-origin by imposing the tumor cell identity giving rise to either B-cell or T-cell leukemias. This novel mechanism of cell transformation is mediated by an epigenetic priming mechanism that is established by the initial genetic lesion. This initial hit might be unnecessary for the subsequent tumor evolution and conservation, being the epigenetic priming the engine for the tumor evolution.
Collapse
Affiliation(s)
- Javier Raboso-Gallego
- Experimental Therapeutics and Translational Oncology Program, Instituto de Biología Molecular y Celular del Cáncer, CSIC/Universidad de Salamanca, Salamanca, Spain.,Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Ana Casado-García
- Experimental Therapeutics and Translational Oncology Program, Instituto de Biología Molecular y Celular del Cáncer, CSIC/Universidad de Salamanca, Salamanca, Spain.,Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Marta Isidro-Hernández
- Experimental Therapeutics and Translational Oncology Program, Instituto de Biología Molecular y Celular del Cáncer, CSIC/Universidad de Salamanca, Salamanca, Spain.,Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | | |
Collapse
|
298
|
Booth C, Romano R, Roncarolo MG, Thrasher AJ. Gene therapy for primary immunodeficiency. Hum Mol Genet 2019; 28:R15-R23. [DOI: 10.1093/hmg/ddz170] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/02/2019] [Accepted: 07/08/2019] [Indexed: 01/21/2023] Open
Abstract
Abstract
Gene therapy is now being trialled as a therapeutic option for an expanding number of conditions, based primarily on the successful treatment over the past two decades of patients with specific primary immunodeficiencies (PIDs) including severe combined immunodeficiency and Wiskott–Aldrich syndrome and metabolic conditions such as leukodystrophy. The field has evolved from the use of gammaretroviral vectors to more sophisticated lentiviral platforms that offer an improved biosafety profile alongside greater efficiency for hematopoietic stem cells gene transfer. Here we review more recent developments including licensing of gene therapies, use of gene corrected autologous T cells as an alternative strategy for some PIDs and the potential of targeted gene correction using various gene editing platforms. Given the promising results of recent clinical trials, it is likely that autologous gene therapies will become standard of care for a number of devastating diseases in the coming decade.
Collapse
Affiliation(s)
- Claire Booth
- Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Rosa Romano
- Division of Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford School of Medicine, Stanford, CA, USA
| | - Maria Grazia Roncarolo
- Division of Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine (ISCBRM), Stanford School of Medicine, Stanford, CA, USA
| | - Adrian J Thrasher
- Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
299
|
Bueren JA, Quintana-Bustamante O, Almarza E, Navarro S, Río P, Segovia JC, Guenechea G. Advances in the gene therapy of monogenic blood cell diseases. Clin Genet 2019; 97:89-102. [PMID: 31231794 DOI: 10.1111/cge.13593] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 05/12/2019] [Accepted: 05/21/2019] [Indexed: 01/19/2023]
Abstract
Hematopoietic gene therapy has markedly progressed during the last 15 years both in terms of safety and efficacy. While a number of serious adverse events (SAE) were initially generated as a consequence of genotoxic insertions of gamma-retroviral vectors in the cell genome, no SAEs and excellent outcomes have been reported in patients infused with autologous hematopoietic stem cells (HSCs) transduced with self-inactivated lentiviral and gammaretroviral vectors. Advances in the field of HSC gene therapy have extended the number of monogenic diseases that can be treated with these approaches. Nowadays, evidence of clinical efficacy has been shown not only in primary immunodeficiencies, but also in other hematopoietic diseases, including beta-thalassemia and sickle cell anemia. In addition to the rapid progression of non-targeted gene therapies in the clinic, new approaches based on gene editing have been developed thanks to the discovery of designed nucleases and improved non-integrative vectors, which have markedly increased the efficacy and specificity of gene targeting to levels compatible with its clinical application. Based on advances achieved in the field of gene therapy, it can be envisaged that these therapies will soon be part of the therapeutic approaches used to treat life-threatening diseases of the hematopoietic system.
Collapse
Affiliation(s)
- Juan A Bueren
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | - Oscar Quintana-Bustamante
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | - Elena Almarza
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | - Susana Navarro
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | - Paula Río
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | - José C Segovia
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | - Guillermo Guenechea
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| |
Collapse
|
300
|
Cooney AL, Singh BK, Loza LM, Thornell IM, Hippee CE, Powers LS, Ostedgaard LS, Meyerholz DK, Wohlford-Lenane C, Stoltz DA, B McCray P, Sinn PL. Widespread airway distribution and short-term phenotypic correction of cystic fibrosis pigs following aerosol delivery of piggyBac/adenovirus. Nucleic Acids Res 2019; 46:9591-9600. [PMID: 30165523 PMCID: PMC6182177 DOI: 10.1093/nar/gky773] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 08/22/2018] [Indexed: 11/14/2022] Open
Abstract
Cystic fibrosis (CF) is a common genetic disease caused by mutations in the gene coding for cystic fibrosis transmembrane conductance regulator (CFTR). Although CF affects multiple organ systems, chronic bacterial infections and inflammation in the lung are the leading causes of morbidity and mortality in people with CF. Complementation with a functional CFTR gene repairs this defect, regardless of the disease-causing mutation. In this study, we used a gene delivery system termed piggyBac/adenovirus (Ad), which combines the delivery efficiency of an adenoviral-based vector with the persistent expression of a DNA transposon-based vector. We aerosolized piggyBac/Ad to the airways of pigs and observed widespread pulmonary distribution of vector. We quantified the regional distribution in the airways and observed transduction of large and small airway epithelial cells of non-CF pigs, with ∼30–50% of surface epithelial cells positive for GFP. We transduced multiple cell types including ciliated, non-ciliated, basal, and submucosal gland cells. In addition, we phenotypically corrected CF pigs following delivery of piggyBac/Ad expressing CFTR as measured by anion channel activity, airway surface liquid pH, and bacterial killing ability. Combining an integrating DNA transposon with adenoviral vector delivery is an efficient method for achieving functional CFTR correction from a single vector administration.
Collapse
Affiliation(s)
- Ashley L Cooney
- Department of Microbiology, The University of Iowa, Iowa City, IA 52242, USA
- Stead Family Department of Pediatrics, The University of Iowa, Iowa City, IA 52242, USA
- Pappajohn Biomedical Institute and the Center for Gene Therapy for Cystic Fibrosis, The University of Iowa, Iowa City, IA 52242, USA
| | - Brajesh K Singh
- Stead Family Department of Pediatrics, The University of Iowa, Iowa City, IA 52242, USA
- Pappajohn Biomedical Institute and the Center for Gene Therapy for Cystic Fibrosis, The University of Iowa, Iowa City, IA 52242, USA
| | - Laura Marquez Loza
- Pappajohn Biomedical Institute and the Center for Gene Therapy for Cystic Fibrosis, The University of Iowa, Iowa City, IA 52242, USA
- Department of Molecular Medicine, The University of Iowa, Iowa City, IA 52242, USA
| | - Ian M Thornell
- Pappajohn Biomedical Institute and the Center for Gene Therapy for Cystic Fibrosis, The University of Iowa, Iowa City, IA 52242, USA
- Department of Internal Medicine, The University of Iowa, Iowa City, IA 52242, USA
| | - Camilla E Hippee
- Stead Family Department of Pediatrics, The University of Iowa, Iowa City, IA 52242, USA
- Pappajohn Biomedical Institute and the Center for Gene Therapy for Cystic Fibrosis, The University of Iowa, Iowa City, IA 52242, USA
| | - Linda S Powers
- Pappajohn Biomedical Institute and the Center for Gene Therapy for Cystic Fibrosis, The University of Iowa, Iowa City, IA 52242, USA
- Department of Internal Medicine, The University of Iowa, Iowa City, IA 52242, USA
| | - Lynda S Ostedgaard
- Pappajohn Biomedical Institute and the Center for Gene Therapy for Cystic Fibrosis, The University of Iowa, Iowa City, IA 52242, USA
- Department of Internal Medicine, The University of Iowa, Iowa City, IA 52242, USA
| | - David K Meyerholz
- Pappajohn Biomedical Institute and the Center for Gene Therapy for Cystic Fibrosis, The University of Iowa, Iowa City, IA 52242, USA
- Department of Pathology, The University of Iowa, Iowa City, IA 52242, USA
| | - Chris Wohlford-Lenane
- Stead Family Department of Pediatrics, The University of Iowa, Iowa City, IA 52242, USA
- Pappajohn Biomedical Institute and the Center for Gene Therapy for Cystic Fibrosis, The University of Iowa, Iowa City, IA 52242, USA
| | - David A Stoltz
- Pappajohn Biomedical Institute and the Center for Gene Therapy for Cystic Fibrosis, The University of Iowa, Iowa City, IA 52242, USA
- Department of Internal Medicine, The University of Iowa, Iowa City, IA 52242, USA
- Department of Pathology, The University of Iowa, Iowa City, IA 52242, USA
| | - Paul B McCray
- Department of Microbiology, The University of Iowa, Iowa City, IA 52242, USA
- Stead Family Department of Pediatrics, The University of Iowa, Iowa City, IA 52242, USA
- Pappajohn Biomedical Institute and the Center for Gene Therapy for Cystic Fibrosis, The University of Iowa, Iowa City, IA 52242, USA
- Department of Molecular Medicine, The University of Iowa, Iowa City, IA 52242, USA
| | - Patrick L Sinn
- Stead Family Department of Pediatrics, The University of Iowa, Iowa City, IA 52242, USA
- Pappajohn Biomedical Institute and the Center for Gene Therapy for Cystic Fibrosis, The University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|